1
|
Haacke N, Wang H, Yan S, Barovic M, Li X, Nagai K, Botezatu A, Hatzioannou A, Gercken B, Trimaglio G, Shah AU, Wang J, Ye L, Jaykar MT, Rauner M, Wielockx B, Chung KJ, Netea MG, Kalafati L, Hajishengallis G, Chavakis T. Innate immune training of osteoclastogenesis promotes inflammatory bone loss in mice. Dev Cell 2025:S1534-5807(25)00063-2. [PMID: 40020679 PMCID: PMC7617534 DOI: 10.1016/j.devcel.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
We previously demonstrated that long-term trained immunity (TRIM) involves adaptations that imprint innate immune memory in long-lived myelopoiesis precursors and their progeny, monocytes/macrophages and neutrophils, which thereby acquire enhanced responsiveness to future challenges. Here, we show that a distinct component of myeloid biology, osteoclastogenesis, can also undergo innate immune training. Indeed, β-glucan-induced TRIM was associated with an increased osteoclastogenesis bias in the bone marrow and an expansion of monocytes/osteoclast progenitors in the periphery, resulting in aggravated severity of experimental periodontitis and arthritis. In the setting of trained inflammatory osteoclastogenesis, we observed transcriptomic rewiring in synovial myeloid cells of arthritic mice, featuring prominent upregulation of the transcription factor melanogenesis-associated transcription factor (MITF). Adoptive transfer of splenic monocytes from β-glucan-trained mice to naive recipients exacerbated arthritis in the latter in a strictly MITF-dependent manner. Our findings establish trained osteoclastogenesis as a maladaptive component of TRIM and potentially provide therapeutic targets in inflammatory bone loss disorders.
Collapse
Affiliation(s)
- Nora Haacke
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shu Yan
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Marko Barovic
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kosuke Nagai
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Adelina Botezatu
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Aikaterini Hatzioannou
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Giulia Trimaglio
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Anisha U Shah
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mangesh T Jaykar
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 XZ Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| |
Collapse
|
2
|
Simons P, Bondu V, Shevy L, Young S, Wandinger-Ness A, Bologa CG, Buranda T. A model for predicting bacteremia species based on host immune response. Front Cell Infect Microbiol 2025; 15:1451293. [PMID: 40041147 PMCID: PMC11876111 DOI: 10.3389/fcimb.2025.1451293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Clinicians encounter significant challenges in quickly and accurately identifying the bacterial species responsible for patient bacteremia and in selecting appropriate antibiotics for timely treatment. This study introduces a novel approach that combines immune response data from routine blood counts with assessments of immune cell activation, specifically through quantitative measurements of Rho family GTPase activity. The combined data were used to develop a machine-learning model capable of distinguishing specific classes of bacteria and their associations. Methods We aimed to determine whether different classes of bacteria elicit distinct patterns of host immune responses, as indicated by quantitative differences in leukocyte populations from routine complete blood counts with differential. Concurrently, we conducted quantitative measurements of activated Rac1 (Rac1•GTP) levels using a novel 'G-Trap assay' we developed. With the G-Trap, we measured Rac1•GTP in peripheral blood monocytes (PBMC) and polymorphonuclear (PMN) cells from blood samples collected from 28 culture-positive patients and over 80 non-infected patients used as controls. Results Our findings indicated that 18 of the 28 patients with bacteremia showed an increase of ≥ 3-fold in Rac1•GTP levels compared to the controls. The remaining ten patients with bacteremia exhibited either neutrophilia or pancytopenia and displayed normal to below-normal Rac1 GTPase activity, which is consistent with bacteria-induced immunosuppression. To analyze the data, we employed partial least squares discriminant analysis (PLS-DA), a supervised method that optimizes group separation and aids in building a novel machine-learning model for pathogen identification. Discussion The results demonstrated that PLS-DA effectively differentiates between specific pathogen groups, and external validation confirmed the predictive model's utility. Given that bacterial culture confirmation may take several days, our study underscores the potential of combining routine assays with a machine-learning model as a valuable clinical decision-support tool. This approach could enable prompt and accurate treatment on the same day that patients present to the clinic.
Collapse
Affiliation(s)
- Peter Simons
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Virginie Bondu
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Laura Shevy
- Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States
| | - Stephen Young
- Tricore Research Laboratories, Albuquerque, NM, United States
| | - Angela Wandinger-Ness
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Cristian G. Bologa
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Tione Buranda
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
3
|
Doye A, Chaintreuil P, Verhoeyen E, Boyer L. [RAC2 gain-of-function mutations causing immunodeficiency drive activation of the NLRP3 inflammasome]. Med Sci (Paris) 2025; 41:133-135. [PMID: 40028950 DOI: 10.1051/medsci/2025008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Affiliation(s)
- Anne Doye
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| | - Paul Chaintreuil
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| | - Els Verhoeyen
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| | - Laurent Boyer
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| |
Collapse
|
4
|
Campbell GP, Amin D, Hsieh K, Hussey GS, St Leger AJ, Gross JM, Badylak SF, Kuwajima T. Immunomodulation by the combination of statin and matrix-bound nanovesicle enhances optic nerve regeneration. NPJ Regen Med 2024; 9:31. [PMID: 39461953 PMCID: PMC11513974 DOI: 10.1038/s41536-024-00374-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
Modulating inflammation is critical to enhance nerve regeneration after injury. However, clinically applicable regenerative therapies that modulate inflammation have not yet been established. Here, we demonstrate synergistic effects of the combination of an HMG-CoA reductase inhibitor, statin/fluvastatin and critical components of the extracellular matrix, Matrix-Bound Nanovesicles (MBV) to enhance axon regeneration and neuroprotection after mouse optic nerve injury. Mechanistically, co-intravitreal injections of fluvastatin and MBV robustly promote infiltration of monocytes and neutrophils, which lead to RGC protection and axon regeneration. Furthermore, monocyte infiltration is triggered by elevated expression of CCL2, a chemokine, in the superficial layer of the retina after treatment with a combination of fluvastatin and MBV or IL-33, a cytokine contained within MBV. Finally, this therapy can be further combined with AAV-based gene therapy blocking anti-regenerative pathways in RGCs to extend regenerated axons. These data highlight novel molecular insights into the development of immunomodulatory regenerative therapy.
Collapse
Affiliation(s)
- Gregory P Campbell
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- The Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Dwarkesh Amin
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- The Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Kristin Hsieh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- The Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - George S Hussey
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Anthony J St Leger
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jeffrey M Gross
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- The Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Takaaki Kuwajima
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
- The Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
5
|
Doye A, Chaintreuil P, Lagresle-Peyrou C, Batistic L, Marion V, Munro P, Loubatier C, Chirara R, Sorel N, Bessot B, Bronnec P, Contenti J, Courjon J, Giordanengo V, Jacquel A, Barbry P, Couralet M, Aladjidi N, Fischer A, Cavazzana M, Mallebranche C, Visvikis O, Kracker S, Moshous D, Verhoeyen E, Boyer L. RAC2 gain-of-function variants causing inborn error of immunity drive NLRP3 inflammasome activation. J Exp Med 2024; 221:e20231562. [PMID: 39212656 PMCID: PMC11363864 DOI: 10.1084/jem.20231562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/15/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
A growing number of patients presenting severe combined immunodeficiencies attributed to monoallelic RAC2 variants have been identified. The expression of the RHO GTPase RAC2 is restricted to the hematopoietic lineage. RAC2 variants have been described to cause immunodeficiencies associated with high frequency of infection, leukopenia, and autoinflammatory features. Here, we show that specific RAC2 activating mutations induce the NLRP3 inflammasome activation leading to the secretion of IL-1β and IL-18 from macrophages. This activation depends on the activation state of the RAC2 variant and is mediated by the downstream kinase PAK1. Inhibiting the RAC2-PAK1-NLRP3 inflammasome pathway might be considered as a potential treatment for these patients.
Collapse
Affiliation(s)
- Anne Doye
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | - Chantal Lagresle-Peyrou
- Université Paris Cité, Paris, France
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | | | | | | | | | - Rayana Chirara
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Nataël Sorel
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Boris Bessot
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Pauline Bronnec
- Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Lyon, France
- Université Claude Bernard Lyon 1, CNRS UMR5308, École normale supérieure de Lyon, Lyon, France
| | - Julie Contenti
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Johan Courjon
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Valerie Giordanengo
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | | | - Pascal Barbry
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Marie Couralet
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Nathalie Aladjidi
- Centre de Référence National des Cytopénies Autoimmunes de l’Enfant, Pediatric Hematologic Unit, Centre d’Investigation Clinique Plurithématique INSERM 1401, University Hospital of Bordeaux, Bordeaux, France
| | - Alain Fischer
- Imagine Institute, INSERM UMR 1163, Paris, France
- Necker Hospital, Pediatric Hematology-Immunology and Rheumatology Unit, Assistance Publique-Hôpitaux de Paris, Paris, France
- Collège de France, Paris, France
| | - Marina Cavazzana
- Université Paris Cité, Paris, France
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Coralie Mallebranche
- Université d’Angers, Université de Nantes, Inserm, CNRS, CRCI2NA, SFR ICAT, Angers, France
- Centre Hospitalier Universitaire Angers, Pediatric Immuno-Hemato-Oncology Unit, Angers, France
| | | | - Sven Kracker
- Université Paris Cité, Paris, France
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Despina Moshous
- Laboratoire Dynamique du Génome et Système Immunitaire, Imagine Institute, INSERM UMR 1163, Paris, France
- Centre de Référence des Déficits Immunitaires Héréditaires, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Els Verhoeyen
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Lyon, France
| | | |
Collapse
|
6
|
Yang L, Xu Q, Li J. Prognostic impact of ARHGAP43(SH3BP1) in acute myeloid leukemia. J Formos Med Assoc 2024; 123:992-1003. [PMID: 38582737 DOI: 10.1016/j.jfma.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy with a heterogeneous prognosis. Novel markers are required to accurately assess the prognosis and formulate treatment plans. METHODS The association of ARHGAP family genes with prognostic value in acute myeloid leukemia (AML) was assessed using public databases (CCLE, GEPIA, TCGA, and GEO). RESULTS Elevated expression of ARHGAP43 (SH3BP1) was associated with poor prognosis in patients with acute myeloid leukemia. ARHGAP43 (SH3BP1) expression was higher in the poor/adverse prognosis (P < 0.001) and TP53 mutation groups (P = 0.0093). Higher ARHGAP43 (SH3BP1) expression was found to be an independent prognostic predictor in multivariate COX regression analysis (HR = 1.317, 95% CI: 1.008-1.720, P = 0.044). Higher ARHGAP43 (SH3BP1) expression who did not receive hematopoietic stem cell transplantation (HSCT) had shorter overall survival (OS) and progression-free survival (PFS) (OS: median: 7.60 vs. 24.90 months; P = 0.006; PFS: median: 11.40 vs. 27.22 months; P = 0.0096), whereas OS and PFS of patients who received HSCT were unaffected, suggesting that HSCT is a better treatment option for patients with higher ARHGAP43 (SH3BP1) expression. KEGG and GSEA analyses revealed that high-expression ARHGAP43 (SH3BP1) was related to inflammation and immune response. Additionally, down-regulation of ARHGAP43 (SH3BP1) expression inhibited AML cell proliferation. CONCLUSION These findings highlight the clinical potential of ARHGAP43 (SH3BP1) as a novel biomarker of AML, with higher levels indicating a poor prognosis.
Collapse
Affiliation(s)
- Li Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qiang Xu
- Department of Orthopedics and Joint Surgery, Qijiang District People's Hospital, Chongqing, 401420, China
| | - Junnan Li
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
7
|
Pandarangga P, Doan PTK, Tearle R, Low WY, Ren Y, Nguyen HTH, Dharmayanti NI, Hemmatzadeh F. mRNA Profiling and Transcriptomics Analysis of Chickens Received Newcastle Disease Virus Genotype II and Genotype VII Vaccines. Pathogens 2024; 13:638. [PMID: 39204239 PMCID: PMC11357267 DOI: 10.3390/pathogens13080638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Newcastle Disease Virus (NDV) genotype VII (GVII) is becoming the predominant strain of NDV in the poultry industry. It causes high mortality even in vaccinated chickens with a common NDV genotype II vaccine (GII-vacc). To overcome this, the killed GVII vaccine has been used to prevent NDV outbreaks. However, the debate about vaccine differences remains ongoing. Hence, this study investigated the difference in chickens' responses to the two vaccines at the molecular level. The spleen transcriptomes from vaccinated chickens reveal that GVII-vacc affected the immune response by downregulating neuroinflammation. It also enhanced a synaptogenesis pathway that operates typically in the nervous system, suggesting a mechanism for the neurotrophic effect of this strain. We speculated that the down-regulated immune system regulation correlated with protecting the nervous system from excess leukocytes and cytokine activity. In contrast, GII-vacc inhibited apoptosis by downregulating PERK/ATF4/CHOP as part of the unfolded protein response pathway but did not affect the expression of the same synaptogenesis pathway. Thus, the application of GVII-vacc needs to be considered in countries where GVII is the leading cause of NDV outbreaks. The predicted molecular signatures may also be used in developing new vaccines that trigger specific genes in the immune system in combating NDV outbreaks.
Collapse
Affiliation(s)
- Putri Pandarangga
- Departemen Klinik, Reproduksi, dan Patologi, Fakultas Kedokteran dan Kedokteran Hewan, Universitas Nusa Cendana, Kupang 85001, Indonesia;
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
| | - Phuong Thi Kim Doan
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
- Department of Veterinary Medicine, Tay Nguyen University, Buon Ma Thuot 630000, Vietnam
| | - Rick Tearle
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (R.T.); (W.Y.L.); (Y.R.)
| | - Wai Yee Low
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (R.T.); (W.Y.L.); (Y.R.)
| | - Yan Ren
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (R.T.); (W.Y.L.); (Y.R.)
| | - Hanh Thi Hong Nguyen
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
| | | | - Farhid Hemmatzadeh
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
| |
Collapse
|
8
|
Velázquez-Vega LE, Rivera-Robles M, Sánchez-Álvarez AO, Vivas-Mejía PE, Aponte-Reyes M, Cruz-Collazo AM, Grafals-Ruiz N, Dorta-Estremera S, Hernández-O'Farrill E, Vlaar CP, Dharmawardhane S. Efficacy and delivery strategies of the dual Rac/Cdc42 inhibitor MBQ-167 in HER2 overexpressing breast cancer. Transl Oncol 2024; 44:101928. [PMID: 38489873 PMCID: PMC10956050 DOI: 10.1016/j.tranon.2024.101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
Trastuzumab and trastuzumab-based treatments are the standard of care for breast cancer patients who overexpress the human epidermal growth factor receptor 2 (HER2). However, patients often develop resistance to trastuzumab via signaling from alternative growth factor receptors that converge to activate guanine nucleotide exchange factors (GEFs) that in turn activate the Rho GTPases Rac and Cdc42. Since Rac and Cdc42 have been implicated in high tumor grade and therapy resistance, inhibiting the activity of Rac and Cdc42 is a rational strategy to overcome HER2-targeted therapy resistance. Therefore, our group developed MBQ-167, a dual Rac/Cdc42 inhibitor with IC50s of 103 nM and 78 nM for Rac and Cdc42, respectively, which is highly effective in reducing cell and tumor growth and metastasis in breast cancer cell and mouse models. Herein, we created a trastuzumab resistant variant of the SKBR3 HER2 positive breast cancer cell line and show that Rac activation is a central mechanism in trastuzumab resistance. Next, we tested the potential of targeting MBQ-167 to HER2 overexpressing trastuzumab-resistant cell lines in vitro, and show that MBQ-167, but not trastuzumab, reduces cell viability and induces apoptosis. When MBQ-167 was targeted to mammary fatpad tumors established from HER2 overexpressing cells via immunoliposomes functionalized with trastuzumab, MBQ-167 and MBQ-167-loaded liposomes show equal efficacy in reducing the viability of trastuzumab-resistant cells, inhibiting tumor growth in mouse xenografts, and reducing metastasis to lungs and liver. This study demonstrates the efficacy of MBQ-167 as an alternative therapeutic in HER2 overexpressing cancers, delivered either in free form or in liposomes.
Collapse
Affiliation(s)
- Luis E Velázquez-Vega
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Michael Rivera-Robles
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | | | - Pablo E Vivas-Mejía
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico; University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | | | - Ailed M Cruz-Collazo
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Nilmary Grafals-Ruiz
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Stephanie Dorta-Estremera
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico; Department of Microbiology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Eliud Hernández-O'Farrill
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Cornelis P Vlaar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Suranganie Dharmawardhane
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico; University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico.
| |
Collapse
|
9
|
Jastrab JB, Kagan JC. Strategies of bacterial detection by inflammasomes. Cell Chem Biol 2024; 31:835-850. [PMID: 38636521 PMCID: PMC11103797 DOI: 10.1016/j.chembiol.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/09/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
Mammalian innate immunity is regulated by pattern-recognition receptors (PRRs) and guard proteins, which use distinct strategies to detect infections. PRRs detect bacterial molecules directly, whereas guards detect host cell manipulations by microbial virulence factors. Despite sensing infection through different mechanisms, both classes of innate immune sensors can activate the inflammasome, an immune complex that can mediate cell death and inflammation. Inflammasome-mediated immune responses are crucial for host defense against many bacterial pathogens and prevent invasion by non-pathogenic organisms. In this review, we discuss the mechanisms by which inflammasomes are stimulated by PRRs and guards during bacterial infection, and the strategies used by virulent bacteria to evade inflammasome-mediated immunity.
Collapse
Affiliation(s)
- Jordan B Jastrab
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jonathan C Kagan
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Szigeti K, Ihnatovych I, Notari E, Dorn RP, Maly I, He M, Birkaya B, Prasad S, Byrne RS, Indurthi DC, Nimmer E, Heo Y, Retfalvi K, Chaves L, Sule N, Hofmann WA, Auerbach A, Wilding G, Bae Y, Reynolds J. CHRFAM7A diversifies human immune adaption through Ca 2+ signalling and actin cytoskeleton reorganization. EBioMedicine 2024; 103:105093. [PMID: 38569318 PMCID: PMC10999709 DOI: 10.1016/j.ebiom.2024.105093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Human restricted genes contribute to human specific traits in the immune system. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of the α7 nicotinic acetylcholine receptor (α7 nAChR), the highest Ca2+ conductor of the ACh receptors implicated in innate immunity. Understanding the mechanism of how CHRFAM7A affects the immune system remains unexplored. METHODS Two model systems are used, human induced pluripotent stem cells (iPSC) and human primary monocytes, to characterize α7 nAChR function, Ca2+ dynamics and decoders to elucidate the pathway from receptor to phenotype. FINDINGS CHRFAM7A/α7 nAChR is identified as a hypomorphic receptor with mitigated Ca2+ influx and prolonged channel closed state. This shifts the Ca2+ reservoir from the extracellular space to the endoplasmic reticulum (ER) leading to Ca2+ dynamic changes. Ca2+ decoder small GTPase Rac1 is then activated, reorganizing the actin cytoskeleton. Observed actin mediated phenotypes include cellular adhesion, motility, phagocytosis and tissue mechanosensation. INTERPRETATION CHRFAM7A introduces an additional, human specific, layer to Ca2+ regulation leading to an innate immune gain of function. Through the actin cytoskeleton it drives adaptation to the mechanical properties of the tissue environment leading to an ability to invade previously immune restricted niches. Human genetic diversity predicts profound translational significance as its understanding builds the foundation for successful treatments for infectious diseases, sepsis, and cancer metastasis. FUNDING This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti) and in part by NIH grant R01HL163168 (Yongho Bae).
Collapse
Affiliation(s)
- Kinga Szigeti
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| | - Ivanna Ihnatovych
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Emily Notari
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ryu P Dorn
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ivan Maly
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Muye He
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Barbara Birkaya
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Shreyas Prasad
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Robin Schwartz Byrne
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Dinesh C Indurthi
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Erik Nimmer
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yuna Heo
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Kolos Retfalvi
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Lee Chaves
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Norbert Sule
- Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY, 14203, USA
| | - Wilma A Hofmann
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Anthony Auerbach
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Gregory Wilding
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yongho Bae
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Jessica Reynolds
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| |
Collapse
|
11
|
Neyton LPA, Sinha P, Sarma A, Mick E, Kalantar K, Chen S, Wu N, Delucchi K, Zhuo H, Bos LDJ, Jauregui A, Gomez A, Hendrickson CM, Kangelaris KN, Leligdowicz A, Liu KD, Matthay MA, Langelier CR, Calfee CS. Host and Microbe Blood Metagenomics Reveals Key Pathways Characterizing Critical Illness Phenotypes. Am J Respir Crit Care Med 2024; 209:805-815. [PMID: 38190719 PMCID: PMC10995577 DOI: 10.1164/rccm.202308-1328oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024] Open
Abstract
Rationale: Two molecular phenotypes of sepsis and acute respiratory distress syndrome, termed hyperinflammatory and hypoinflammatory, have been consistently identified by latent class analysis in numerous cohorts, with widely divergent clinical outcomes and differential responses to some treatments; however, the key biological differences between these phenotypes remain poorly understood.Objectives: We used host and microbe metagenomic sequencing data from blood to deepen our understanding of biological differences between latent class analysis-derived phenotypes and to assess concordance between the latent class analysis-derived phenotypes and phenotypes reported by other investigative groups (e.g., Sepsis Response Signature [SRS1-2], molecular diagnosis and risk stratification of sepsis [MARS1-4], reactive and uninflamed).Methods: We analyzed data from 113 patients with hypoinflammatory sepsis and 76 patients with hyperinflammatory sepsis enrolled in a two-hospital prospective cohort study. Molecular phenotypes had been previously assigned using latent class analysis.Measurements and Main Results: The hyperinflammatory and hypoinflammatory phenotypes of sepsis had distinct gene expression signatures, with 5,755 genes (31%) differentially expressed. The hyperinflammatory phenotype was associated with elevated expression of innate immune response genes, whereas the hypoinflammatory phenotype was associated with elevated expression of adaptive immune response genes and, notably, T cell response genes. Plasma metagenomic analysis identified differences in prevalence of bacteremia, bacterial DNA abundance, and composition between the phenotypes, with an increased presence and abundance of Enterobacteriaceae in the hyperinflammatory phenotype. Significant overlap was observed between these phenotypes and previously identified transcriptional subtypes of acute respiratory distress syndrome (reactive and uninflamed) and sepsis (SRS1-2). Analysis of data from the VANISH trial indicated that corticosteroids might have a detrimental effect in patients with the hypoinflammatory phenotype.Conclusions: The hyperinflammatory and hypoinflammatory phenotypes have distinct transcriptional and metagenomic features that could be leveraged for precision treatment strategies.
Collapse
Affiliation(s)
| | - Pratik Sinha
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine
| | - Eran Mick
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine
- Division of Infectious Diseases
- Chan Zuckerberg Biohub, San Francisco, California
| | | | | | - Nelson Wu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine
| | | | | | - Lieuwe D J Bos
- Department of Intensive Care and Laboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Antonio Gomez
- Department of Medicine
- Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California; and
| | - Carolyn M Hendrickson
- Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California; and
| | | | | | | | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - Charles R Langelier
- Division of Infectious Diseases
- Chan Zuckerberg Biohub, San Francisco, California
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine
- Department of Anesthesiology, and
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| |
Collapse
|
12
|
Zhang S, Zhang L, Liu T, Qiao Y, Cao X, Cheng J, Wu H, Shen H. Investigating the transcriptomic variances in two phases Ecytonucleospora hepatopenaei (EHP) in Litopenaeus vannamei. J Invertebr Pathol 2024; 203:108061. [PMID: 38244837 DOI: 10.1016/j.jip.2024.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
This study explores the transcriptomic differences in two distinct phases of Ecytonucleospora hepatopenaei (EHP) in Litopenaeus vannamei, a crucial aspect in shrimp health management. We employed high-throughput sequencing to categorize samples into two phases, 'Phase A' and 'Phase B', defined by the differential expression of PTP2 and TPS1 genes. Our analysis identified 2057 genes, with 78 exhibiting significant variances, including 62 upregulated and 16 downregulated genes. Enrichment analyses via GO and KEGG pathways highlighted these genes' roles in cellular metabolism, signal transduction, and immune responses. Notably, genes like IQGAP2, Rhob, Pim1, and PCM1 emerged as potentially crucial in EHP's infection process and lifecycle. We hypothesize that these genes may influence trehalose metabolism and glucose provision, impacting the biological activities within EHP during different phases. Interestingly, a lower transcript count in 'Phase A' EHP suggests a reduction in biological activities, likely preparing for host cell invasion. This research provides a foundational understanding of EHP infection mechanisms, offering vital insights for future studies and therapeutic interventions.
Collapse
Affiliation(s)
- Sheng Zhang
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China; Jiangsu Ocean University, Lianyungang 222005, China
| | - Leiting Zhang
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China; Nanjing Normal University, Nanjing 210023, China
| | - Tingyue Liu
- Nanjing Normal University, Nanjing 210023, China
| | - Yi Qiao
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China
| | - Xiaohui Cao
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China
| | - Jie Cheng
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China
| | - Hailong Wu
- Jiangsu Ocean University, Lianyungang 222005, China
| | - Hui Shen
- Jiangsu Marine Fisheries Research Institute, Nantong 226007, China; Jiangsu Ocean University, Lianyungang 222005, China; Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
13
|
Tanner CD, Rosowski EE. Macrophages inhibit extracellular hyphal growth of A. fumigatus through Rac2 GTPase signaling. Infect Immun 2024; 92:e0038023. [PMID: 38168666 PMCID: PMC10863406 DOI: 10.1128/iai.00380-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Macrophages act as a first line of defense against pathogens. Against Aspergillus fumigatus, a fungus with pathogenic potential in immunocompromised patients, macrophages can phagocytose fungal spores and inhibit spore germination to prevent the development of tissue-invasive hyphae. However, the cellular pathways that macrophages use to accomplish these tasks and any roles macrophages have later in infection against invasive forms of fungi are still not fully known. Rac-family Rho GTPases are signaling hubs for multiple cellular functions in leukocytes, including cell migration, phagocytosis, reactive oxygen species (ROS) generation, and transcriptional activation. We therefore aimed to further characterize the function of macrophages against A. fumigatus in an in vivo vertebrate infection model by live imaging of the macrophage behavior in A. fumigatus-infected rac2 mutant zebrafish larvae. While Rac2-deficient zebrafish larvae are susceptible to A. fumigatus infection, Rac2 deficiency does not impair macrophage migration to the infection site, interaction with and phagocytosis of spores, spore trafficking to acidified compartments, or spore killing. However, we reveal a role for Rac2 in macrophage-mediated inhibition of spore germination and control of invasive hyphae. Re-expression of Rac2 under a macrophage-specific promoter rescues the survival of A. fumigatus-infected rac2 mutant larvae through increased control of germination and hyphal growth. Altogether, we describe a new role for macrophages against extracellular hyphal growth of A. fumigatus and report that the function of the Rac2 Rho GTPase in macrophages is required for this function.
Collapse
Affiliation(s)
- Christopher D. Tanner
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Emily E. Rosowski
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
14
|
Mishra AK, Rodriguez M, Torres AY, Smith M, Rodriguez A, Bond A, Morrissey MA, Montell DJ. Hyperactive Rac stimulates cannibalism of living target cells and enhances CAR-M-mediated cancer cell killing. Proc Natl Acad Sci U S A 2023; 120:e2310221120. [PMID: 38109551 PMCID: PMC10756302 DOI: 10.1073/pnas.2310221120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/15/2023] [Indexed: 12/20/2023] Open
Abstract
The 21kD GTPase Rac is an evolutionarily ancient regulator of cell shape and behavior. Rac2 is predominantly expressed in hematopoietic cells where it is essential for survival and motility. The hyperactivating mutation Rac2E62K also causes human immunodeficiency, although the mechanism remains unexplained. Here, we report that in Drosophila, hyperactivating Rac stimulates ovarian cells to cannibalize neighboring cells, destroying the tissue. We then show that hyperactive Rac2E62K stimulates human HL60-derived macrophage-like cells to engulf and kill living T cell leukemia cells. Primary mouse Rac2+/E62K bone-marrow-derived macrophages also cannibalize primary Rac2+/E62K T cells due to a combination of macrophage hyperactivity and T cell hypersensitivity to engulfment. Additionally, Rac2+/E62K macrophages non-autonomously stimulate wild-type macrophages to engulf T cells. Rac2E62K also enhances engulfment of target cancer cells by chimeric antigen receptor-expressing macrophages (CAR-M) in a CAR-dependent manner. We propose that Rac-mediated cell cannibalism may contribute to Rac2+/E62K human immunodeficiency and enhance CAR-M cancer immunotherapy.
Collapse
Affiliation(s)
- Abhinava K. Mishra
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Melanie Rodriguez
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Alba Yurani Torres
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Morgan Smith
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Anthony Rodriguez
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Meghan A. Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| | - Denise J. Montell
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106
| |
Collapse
|
15
|
Hady TF, Hwang B, Waworuntu RL, Ratner BD, Bryers JD. Cells resident to precision templated 40-µm pore scaffolds generate small extracellular vesicles that affect CD4 + T cell phenotypes through regulatory TLR4 signaling. Acta Biomater 2023; 166:119-132. [PMID: 37150279 PMCID: PMC10330460 DOI: 10.1016/j.actbio.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Precision porous templated scaffolds (PTS) are a hydrogel construct of uniformly sized interconnected spherical pores that induce a pro-healing response (reducing the foreign body reaction, FBR) exclusively when the pores are 30-40µm in diameter. Our previous work demonstrated the necessity of Tregs in the maintenance of PTS pore size specific differences in CD4+ T cell phenotype. Work here characterizes the role of Tregs in the responses to implanted 40µm and 100µm PTS using WT and FoxP3+ cell (Treg) depleted mice. Proteomic analyses indicate that integrin signaling, monocytes/macrophages, cytoskeletal remodeling, inflammatory cues, and vesicule endocytosis may participate in Treg activation and the CD4+ T cell equilibrium modulated by PTS resident cell-derived small extracellular vesicles (sEVs). The role of MyD88-dependent and MyD88-independent TLR4 activation in PTS cell-derived sEV-to-T cell signaling is quantified by treating WT, TLR4ko, and MyD88ko splenic T cells with PTS cell-derived sEVs. STAT3 and mTOR are identified as mechanisms for further study for pore-size dependent PTS cell-derived sEV-to-T cell signaling. STATEMENT OF SIGNIFICANCE: Unique cell populations colonizing only within 40µm pore size PTS generate sEVs that resolve inflammation by modifying CD4+ T cell phenotypes through TLR4 signaling.
Collapse
Affiliation(s)
- T F Hady
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - B Hwang
- Center for Lung Biology, Department of Surgery, University of Washington Seattle, WA 98109, USA
| | - R L Waworuntu
- Center for Lung Biology, Department of Surgery, University of Washington Seattle, WA 98109, USA
| | - B D Ratner
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - J D Bryers
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
16
|
Zhang L, Lv G, Peng Y, Yang L, Chen J, An Y, Zhang Z, Tang X, Li Z, Zhao X. A Novel RAC2 Mutation Causing Combined Immunodeficiency. J Clin Immunol 2023; 43:229-240. [PMID: 36190591 DOI: 10.1007/s10875-022-01373-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/24/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE Ras-related C3 botulinum toxin substrate 2 (RAC2) acts as a molecular switch and has crucial roles in cell signaling and actin dynamics. A broad spectrum of genetic RAC2 mutations can cause various types of primary immunodeficiency, with complete penetrance. Here, we report a novel heterozygous missense mutation in RAC2 with incomplete penetrance, and the associated phenotypes, in a Chinese family. METHODS Immunological phenotype was detected by flow cytometry. T cell receptor excision circles (TRECs) and K-deleting recombination excision circles (KRECs) were assessed by real-time quantitative PCR. Gene mutations were detected by whole-exome sequencing (WES) and confirmed by Sanger sequencing. RESULTS The proband was an 11-year-old girl who presented with recurrent respiratory infections, bronchiectasis, persistent Epstein-Barr virus viremia, infectious mononucleosis, encephalitis, and cutaneous human papillomavirus infections. Laboratory analyses revealed increased serum IgG and decreased IgM levels, reduced naïve CD4+ and CD8+ T cells, an inverted CD4+/CD8+ ratio, and low TREC and KREC numbers. The mutation resulted in increased production of reactive oxygen species, while impaired actin polarization in neutrophils; diminished proliferative responses, increased cytokine production and a dysregulated phenotype in T lymphocytes; as well as accelerated apoptosis and hyperactivity of AKT in HL-60 human leukemia cells. WES identified a c.44G > A mutation in RAC2 resulting in a p.G15D substitution. Despite sharing the same mutation as the proband, her father suffered from recurrent respiratory infections and bronchiectasis, and had similar immunological defects, whereas her sister was apparently healthy, other than cutaneous human papillomavirus infections, and only mild immunological defects were detected preliminarily. CONCLUSIONS Our findings broaden the clinical and genetic spectra of RAC2 mutations and underline the importance of RAC2 gain-of-function mutations with complete or incomplete penetrance.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, Changsha, Hunan, China
- The School of Pediatrics, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ge Lv
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Peng
- The School of Pediatrics, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Pediatrics Research Institute, Hunan Children's Hospital, Changsha, Hunan, China
| | - Lu Yang
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Junjie Chen
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yunfei An
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhang
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Tang
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihui Li
- Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, Changsha, Hunan, China.
- The School of Pediatrics, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| | - Xiaodong Zhao
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Division of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
17
|
Wisniewski É, Czárán D, Kovács F, Bahurek E, Németh A, Sasvári P, Szanda G, Pettkó-Szandtner A, Klement E, Ligeti E, Csépányi-Kömi R. A novel BRET-Based GAP assay reveals phosphorylation-dependent regulation of the RAC-specific GTPase activating protein ARHGAP25. FASEB J 2022; 36:e22584. [PMID: 36190314 DOI: 10.1096/fj.202200689r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
ARHGAP25, a RAC-specific GTPase activating protein (GAP), is an essential regulator of phagocyte effector functions such as phagocytosis, superoxide production, and transendothelial migration. Furthermore, its complex role in tumor behavior has recently been recognized. We previously demonstrated that phosphorylation of serine 363 in ARHGAP25 regulates hematopoietic stem cells and progenitor cells in mouse bone marrow. However, the significance of other potential phosphorylation sites of ARHGAP25 remained unknown. Now, we developed a novel, real-time bioluminescence resonance energy transfer (BRET) assay to monitor the GAP activity of ARHGAP25 in vitro. Using this approach, we revealed that phosphorylation of S363 and S488, but not that of S379-380, controls ARHGAP25's RACGAP activity. On the other hand, we found in granulocyte-differentiated human PLB-985 cells that superoxide production and actin depolymerization are regulated by residues S363 and S379-380. The present data demonstrate the value of our BRET-GAP assay and show that different phosphorylation patterns regulate ARHGAP25's GAP activity and its effect on superoxide production and phagocytosis.
Collapse
Affiliation(s)
- Éva Wisniewski
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Domonkos Czárán
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Fanni Kovács
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Enikő Bahurek
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Afrodité Németh
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Sasvári
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gergő Szanda
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Eva Klement
- Laboratory of Proteomics Research, Biological Research Centre, Szeged, Hungary.,Single Cell Omics ACF, Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
18
|
Ramachandran RP, Nandi I, Haritan N, Zlotkin-Rivkin E, Keren Y, Danieli T, Lebendiker M, Melamed-Book N, Breuer W, Reichmann D, Aroeti B. EspH interacts with the host active Bcr related (ABR) protein to suppress RhoGTPases. Gut Microbes 2022; 14:2130657. [PMID: 36219160 PMCID: PMC9559323 DOI: 10.1080/19490976.2022.2130657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Enteropathogenic Escherichia coli are bacterial pathogens that colonize the gut and cause severe diarrhea in humans. Upon intimate attachment to the intestinal epithelium, these pathogens translocate via a type III secretion system virulent proteins, termed effectors, into the host cells. These effectors manipulate diverse host cell organelles and functions for the pathogen's benefit. However, the precise mechanisms underlying their activities are not fully understood despite intensive research. EspH, a critical effector protein, has been previously reported to disrupt the host cell actin cytoskeleton by suppressing RhoGTPase guanine exchange factors. However, native host proteins targeted by EspH to mediate these activities remained unknown. Here, we identified the active Bcr related (ABR), a protein previously characterized to possess dual Rho guanine nucleotide exchange factor and GTPase activating protein (GAP) domains, as a native EspH interacting partner. These interactions are mediated by the effector protein's C-terminal 38 amino acid segment. The effector primarily targets the GAP domain of ABR to suppress Rac1 and Cdc42, host cell cytotoxicity, bacterial invasion, and filopodium formation at infection sites. Knockdown of ABR expression abolished the ability of EspH to suppress Rac1, Cdc42. Our studies unravel a novel mechanism by which host RhoGTPases are hijacked by bacterial effectors.
Collapse
Affiliation(s)
- Rachana Pattani Ramachandran
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ipsita Nandi
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nir Haritan
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Efrat Zlotkin-Rivkin
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yael Keren
- The Protein Production Facility, Wolfson Centre for Applied Structural Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tsafi Danieli
- The Protein Production Facility, Wolfson Centre for Applied Structural Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mario Lebendiker
- The Protein Production Facility, Wolfson Centre for Applied Structural Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Naomi Melamed-Book
- Bioimaging Unit, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - William Breuer
- Proteomics and Mass Spectrometry Unit, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dana Reichmann
- Proteomics and Mass Spectrometry Unit, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel,Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benjamin Aroeti
- Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel,CONTACT Benjamin Aroeti Department of Cell and Developmental Biology, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem91904, Israel
| |
Collapse
|
19
|
Consalvo KM, Kirolos SA, Sestak CE, Gomer RH. Sex-Based Differences in Human Neutrophil Chemorepulsion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:354-367. [PMID: 35793910 PMCID: PMC9283293 DOI: 10.4049/jimmunol.2101103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/02/2022] [Indexed: 05/25/2023]
Abstract
A considerable amount is known about how eukaryotic cells move toward an attractant, and the mechanisms are conserved from Dictyostelium discoideum to human neutrophils. Relatively little is known about chemorepulsion, where cells move away from a repellent signal. We previously identified pathways mediating chemorepulsion in Dictyostelium, and here we show that these pathways, including Ras, Rac, protein kinase C, PTEN, and ERK1 and 2, are required for human neutrophil chemorepulsion, and, as with Dictyostelium chemorepulsion, PI3K and phospholipase C are not necessary, suggesting that eukaryotic chemorepulsion mechanisms are conserved. Surprisingly, there were differences between male and female neutrophils. Inhibition of Rho-associated kinases or Cdc42 caused male neutrophils to be more repelled by a chemorepellent and female neutrophils to be attracted to the chemorepellent. In the presence of a chemorepellent, compared with male neutrophils, female neutrophils showed a reduced percentage of repelled neutrophils, greater persistence of movement, more adhesion, less accumulation of PI(3,4,5)P3, and less polymerization of actin. Five proteins associated with chemorepulsion pathways are differentially abundant, with three of the five showing sex dimorphism in protein localization in unstimulated male and female neutrophils. Together, this indicates a fundamental difference in a motility mechanism in the innate immune system in men and women.
Collapse
Affiliation(s)
| | - Sara A Kirolos
- Department of Biology, Texas A&M University, College Station, TX
| | - Chelsea E Sestak
- Department of Biology, Texas A&M University, College Station, TX
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX
| |
Collapse
|
20
|
Guo J, Ning Y, Su Z, Guo L, Gu Y. Identification of hub genes and regulatory networks in histologically unstable carotid atherosclerotic plaque by bioinformatics analysis. BMC Med Genomics 2022; 15:145. [PMID: 35773742 PMCID: PMC9245266 DOI: 10.1186/s12920-022-01257-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/28/2022] [Indexed: 01/01/2023] Open
Abstract
Objective This study identified underlying genetic molecules associated with histologically unstable carotid atherosclerotic plaques through bioinformatics analysis that may be potential biomarkers and therapeutic targets. Methods Three transcriptome datasets (GSE41571, GSE120521 and E-MTAB-2055) and one non-coding RNA dataset (GSE111794) that met histological grouping criteria of unstable plaque were downloaded. The common differentially expressed genes (co-DEGs) of unstable plaques identified from three mRNA datasets were annotated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomics (KEGG). A protein–protein interaction (PPI) network was constructed to present the interaction between co-DEGs and screen out hub genes. MiRNet database and GSE111794 dataset were used to identify the miRNAs targeting hub genes. Associated transcription factors (TFs) and drugs were also predicted. These predicted results were used to construct miRNA/TFs-hub gene and drug-hub gene regulatory networks. Results A total of 105 co-DEGs were identified, including 42 up-regulated genes and 63 down-regulated genes, which were mainly enriched in collagen-containing extracellular matrix, focal adhesion, actin filament bundle, chemokine signaling pathway and regulates of actin cytoskeleton. Ten hub genes (up-regulated: HCK, C1QC, CD14, FCER1G, LCP1 and RAC2; down-regulated: TPM1, MYH10, PLS3 and FMOD) were screened. HCK and RAC2 were involved in chemokine signaling pathway, MYH10 and RAC2 were involved in regulation of actin cytoskeleton. We also predicted 12 miRNAs, top5 TFs and 25 drugs targeting hub genes. In the miRNA/TF-hub gene regulatory network, PLS3 was the most connected hub genes and was targeted by six miRNAs and all five screened TFs. In the drug-hub gene regulatory network, HCK was targeted by 20 drugs including 10 inhibitors. Conclusions We screened 10 hub genes and predicted miRNAs and TFs targeting them. These molecules may play a crucial role in the progression of histologically unstable carotid plaques and serve as potential biomarkers and therapeutic targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01257-1.
Collapse
Affiliation(s)
- Julong Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yachan Ning
- Department of Intensive Care Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhixiang Su
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Lianrui Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
21
|
Small AG, Perveen K, Putty T, Patel N, Quinn P, Wechalekar MD, Hii CS, Quach A, Ferrante A. Neutrophils Require Activation to Express Functional Cell-Surface Complement Receptor Immunoglobulin. Front Immunol 2022; 13:840510. [PMID: 35317169 PMCID: PMC8934411 DOI: 10.3389/fimmu.2022.840510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
The phagocytosis-promoting complement receptor, Complement Receptor Immunoglobulin (CRIg), is exclusively expressed on macrophages. It has been demonstrated that expression in macrophages could be modulated by inflammatory mediators, including cytokines. This raised the possibility that a major phagocyte, the neutrophil, may also express CRIg following activation with inflammatory mediators. Here we show that resting peripheral blood neutrophil lysates subjected to protein analysis by Western blot revealed a 35 kDa CRIg isoform, consistent with the expression of CRIg mRNA by RT-PCR. By flow cytometry, CRIg was detected intracellularly and in very minor amounts on the cell surface. Interestingly, expression on the cell surface was significantly increased to functional levels after activation with inflammatory mediators/neutrophil activators; N-Formylmethionine-leucyl-phenylalanine, tumor necrosis factor (TNF), Granulocyte-Macrophage Colony stimulating Factor (GM-CSF), bacterial lipopolysaccharide, leukotriene B4 and phorbol myristate acetate. The increase in expression required p38 MAP kinase and protein kinase C activation, as well as intracellular calcium. Neutrophils which were defective in actin microfilament reorganization due to a mutation in ARPC1B or inhibition of its upstream regulator, Rac2 lose their ability to upregulate CRIg expression. Inhibition of another small GTPase, Rab27a, with pharmacological inhibitors prevented the increase in CRIg expression, suggesting a requirement for the actin cytoskeleton and exocytosis. Engagement of CRIg on TNF-primed neutrophils with an anti-CRIg monoclonal antibody increased the release of superoxide and promoted the activation of p38 but not ERK1/ERK2 or JNK MAP kinases. The TNF-induced increase in killing of Staphylococcus aureus was blocked by the anti-CRIg antibody. Adding to the anti-microbial role of CRIg, it was found that GM-CSF priming lead to the release of neutrophil extracellular traps. Interestingly in contrast to the above mediators the anti-inflammatory cytokine IL-10 caused a decrease in basal expression and GM-CSF induced increase in CRIg expression. The data demonstrate that neutrophils also express CRIg which is regulated by inflammatory mediators and cytokines. The findings show that the neutrophil antimicrobial function involving CRIg requires priming as a means of arming the cell strategically with microbial invasion of tissues and the bloodstream.
Collapse
Affiliation(s)
- Annabelle G. Small
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Rheumatology Department, College of Medicine and Public Health, Flinders Medical Centre, Flinders University, Bedfort Park, SA, Australia
| | - Khalida Perveen
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Trishni Putty
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Nikita Patel
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Patrick Quinn
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Department of Allergy and Immunology, Women’s and Children’s Health Network, North Adelaide, SA, Australia
| | - Mihir D. Wechalekar
- Rheumatology Department, College of Medicine and Public Health, Flinders Medical Centre, Flinders University, Bedfort Park, SA, Australia
| | - Charles S. Hii
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alex Quach
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Antonio Ferrante
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Department of Molecular and Biomedical Science, School of Biological Sciences, University of Adelaide, North Adelaide, SA, Australia
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Antonio Ferrante,
| |
Collapse
|
22
|
Nath AS, Parsons BD, Makdissi S, Chilvers RL, Mu Y, Weaver CM, Euodia I, Fitze KA, Long J, Scur M, Mackenzie DP, Makrigiannis AP, Pichaud N, Boudreau LH, Simmonds AJ, Webber CA, Derfalvi B, Hammon Y, Rachubinski RA, Di Cara F. Modulation of the cell membrane lipid milieu by peroxisomal β-oxidation induces Rho1 signaling to trigger inflammatory responses. Cell Rep 2022; 38:110433. [PMID: 35235794 DOI: 10.1016/j.celrep.2022.110433] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/21/2021] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
Phagocytosis, signal transduction, and inflammatory responses require changes in lipid metabolism. Peroxisomes have key roles in fatty acid homeostasis and in regulating immune function. We find that Drosophila macrophages lacking peroxisomes have perturbed lipid profiles, which reduce host survival after infection. Using lipidomic, transcriptomic, and genetic screens, we determine that peroxisomes contribute to the cell membrane glycerophospholipid composition necessary to induce Rho1-dependent signals, which drive cytoskeletal remodeling during macrophage activation. Loss of peroxisome function increases membrane phosphatidic acid (PA) and recruits RhoGAPp190 during infection, inhibiting Rho1-mediated responses. Peroxisome-glycerophospholipid-Rho1 signaling also controls cytoskeleton remodeling in mouse immune cells. While high levels of PA in cells without peroxisomes inhibit inflammatory phenotypes, large numbers of peroxisomes and low amounts of cell membrane PA are features of immune cells from patients with inflammatory Kawasaki disease and juvenile idiopathic arthritis. Our findings reveal potential metabolic markers and therapeutic targets for immune diseases and metabolic disorders.
Collapse
Affiliation(s)
- Anu S Nath
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Brendon D Parsons
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Stephanie Makdissi
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Rebecca L Chilvers
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Yizhu Mu
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Ceileigh M Weaver
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Irene Euodia
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Katherine A Fitze
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Juyang Long
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Michal Scur
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Duncan P Mackenzie
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Andrew P Makrigiannis
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Nicolas Pichaud
- Université de Moncton, Department of Chemistry and Biochemistry, Moncton, NB E1A 3E9, Canada; New Brunswick Centre for Precision Medicine (NBCPM), Moncton, NB E1A 3E9, Canada
| | - Luc H Boudreau
- Université de Moncton, Department of Chemistry and Biochemistry, Moncton, NB E1A 3E9, Canada; New Brunswick Centre for Precision Medicine (NBCPM), Moncton, NB E1A 3E9, Canada
| | - Andrew J Simmonds
- University of Alberta, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Christine A Webber
- University of Alberta, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Beata Derfalvi
- Dalhousie University, Department of Pediatrics, Halifax, NS B3K 6R8, Canada
| | - Yannick Hammon
- INSERM au Centre d'Immunologie de Marseille Luminy, Marseille 13288, France
| | | | - Francesca Di Cara
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada; Dalhousie University, Department of Pediatrics, Halifax, NS B3K 6R8, Canada.
| |
Collapse
|
23
|
Weidhaas J, Marco N, Scheffler AW, Kalbasi A, Wilenius K, Rietdorf E, Gill J, Heilig M, Desler C, Chin RK, Kaprealian T, McCloskey S, Raldow A, Raja NP, Kesari S, Carrillo J, Drakaki A, Scholz M, Telesca D. Germline biomarkers predict toxicity to anti-PD1/PDL1 checkpoint therapy. J Immunother Cancer 2022; 10:jitc-2021-003625. [PMID: 35115362 PMCID: PMC8804679 DOI: 10.1136/jitc-2021-003625] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND There is great interest in finding ways to identify patients who will develop toxicity to cancer therapies. This has become especially pressing in the era of immune therapy, where toxicity can be long-lasting and life-altering, and primarily comes in the form of immune-related adverse effects (irAEs). Treatment with the first drugs in this class, anti-programmed death 1 (anti-PD1)/programmed death-ligand 1 (PDL1) checkpoint therapies, results in grade 2 or higher irAEs in up to 25%-30% of patients, which occur most commonly within the first 6 months of treatment and can include arthralgias, rash, pruritus, pneumonitis, diarrhea and/or colitis, hepatitis, and endocrinopathies. We tested the hypothesis that germline microRNA pathway functional variants, known to predict altered systemic stress responses to cancer therapies, would predict irAEs in patients across cancer types. METHODS MicroRNA pathway variants were evaluated for an association with grade 2 or higher toxicity using four classifiers on 62 patients with melanoma, and then the panel's performance was validated on 99 patients with other cancer types. Trained classifiers included classification trees, LASSO-regularized logistic regression, boosted trees, and random forests. Final performance measures were reported on the training set using leave-one-out cross validation and validated on held-out samples. The predicted probability of toxicity was evaluated for its association, if any, with response categories to anti-PD1/PDL1 therapy in the melanoma cohort. RESULTS A biomarker panel was identified that predicts toxicity with 80% accuracy (F1=0.76, area under the curve (AUC)=0.82) in the melanoma training cohort and 77.6% accuracy (F1=0.621, AUC=0.778) in the pan-cancer validation cohort. In the melanoma cohort, the predictive probability of toxicity was not associated with response categories to anti-PD1/PDL1 therapy (p=0.70). In the same cohort, the most significant biomarker of toxicity in RAC1, predicting a greater than ninefold increased risk of toxicity (p<0.001), was also not associated with response to anti-PD1/PDL1 therapy (p=0.151). CONCLUSIONS A germline microRNA-based biomarker signature predicts grade 2 and higher irAEs to anti-PD1/PDL1 therapy, regardless of tumor type, in a pan-cancer manner. These findings represent an important step toward personalizing checkpoint therapy, the use of which is growing rapidly.
Collapse
Affiliation(s)
- Joanne Weidhaas
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Nicholas Marco
- Department of Biostatistics, UCLA, Los Angeles, California, USA
| | | | - Anusha Kalbasi
- Department of Biostatistics, UCLA, Los Angeles, California, USA
| | - Kirk Wilenius
- Prostate Oncology Specialists, Marina Del Rey, California, USA
| | - Emily Rietdorf
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Jaya Gill
- Pacific Neuroscience Institute and Saint John’s Cancer Institute, Santa Monica, California, USA
| | - Mara Heilig
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Caroline Desler
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Robert K Chin
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Tania Kaprealian
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Susan McCloskey
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Ann Raldow
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Naga P Raja
- Appalachian Regional Healthcare, Hazard, Kentucky, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute and Saint John’s Cancer Institute, Santa Monica, California, USA
| | - Jose Carrillo
- Pacific Neuroscience Institute and Saint John’s Cancer Institute, Santa Monica, California, USA
| | - Alexandra Drakaki
- Department of Urology, Medical Oncology, University of California Los Angeles, Los Angeles, California, USA
| | - Mark Scholz
- Prostate Oncology Specialists, Marina Del Rey, California, USA
| | | |
Collapse
|
24
|
Kim H, Takegahara N, Choi Y. Protocadherin-7 Regulates Osteoclast Differentiation through Intracellular SET-Binding Domain-Mediated RhoA and Rac1 Activation. Int J Mol Sci 2021; 22:13117. [PMID: 34884920 PMCID: PMC8658210 DOI: 10.3390/ijms222313117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022] Open
Abstract
Protocadherin-7 (Pcdh7) is a member of the non-clustered protocadherin δ1 subgroup of the cadherin superfamily. Although the cell-intrinsic role of Pcdh7 in osteoclast differentiation has been demonstrated, the molecular mechanisms of Pcdh7 regulating osteoclast differentiation remain to be determined. Here, we demonstrate that Pcdh7 contributes to osteoclast differentiation by regulating small GTPases, RhoA and Rac1, through its SET oncoprotein binding domain. Pcdh7 is associated with SET along with RhoA and Rac1 during osteoclast differentiation. Pcdh7-deficient (Pcdh7-/-) cells showed abolished RANKL-induced RhoA and Rac1 activation, and impaired osteoclast differentiation. Impaired osteoclast differentiation in Pcdh7-/- cells was restored by retroviral transduction of full-length Pcdh7 but not by a Pcdh7 mutant that lacks SET binding domain. The direct crosslink of the Pcdh7 intracellular region induced the activation of RhoA and Rac1, which was not observed when Pcdh7 lacks the SET binding domain. Additionally, retroviral transduction of the constitutively active form of RhoA and Rac1 completely restored the impaired osteoclast differentiation in Pcdh7-/- cells. Collectively, these results demonstrate that Pcdh7 controls osteoclast differentiation by regulating RhoA and Rac1 activation through the SET binding domain.
Collapse
Affiliation(s)
| | | | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.K.); (N.T.)
| |
Collapse
|
25
|
de la Ballina NR, Villalba A, Cao A. Shotgun analysis to identify differences in protein expression between granulocytes and hyalinocytes of the European flat oyster Ostrea edulis. FISH & SHELLFISH IMMUNOLOGY 2021; 119:678-691. [PMID: 34748932 DOI: 10.1016/j.fsi.2021.10.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 10/19/2021] [Accepted: 10/31/2021] [Indexed: 06/13/2023]
Abstract
Recovery of wild populations of the European flat oyster Ostrea edulis is important for ecosystem health and conservation of this species, because native oyster populations have dramatically declined or disappeared in most European waters. Diseases have contributed to oyster decline and are important constrains for oyster recovery. Understanding oyster immune system should contribute to design effective strategies to fight oyster diseases. Haemocytes play a pivotal role in mollusc immune responses protecting from infection. Two main types of haemocytes, granulocytes and hyalinocytes, are distinguished in O. edulis. A study aiming to explore differential functions between both haemocyte types and, thus, to enrich the knowledge of Ostrea edulis immune system, was performed by comparing the proteome of the two haemolymph cell types, using a shotgun approach through liquid chromatography (LC) coupled to mass spectrometry (MS). Cells from oyster haemolymph were differentially separated by Percoll density gradient centrifugation. Shotgun LC-MS/MS performance allowed the identification of 145 proteins in hyalinocytes and 138 in the proteome of granulocytes. After a comparative analysis, 55 proteins with main roles in defence were identified, from which 28 were representative of granulocytes and 27 of hyalinocytes, plus 11 proteins shared by both cell types. Different proteins involved in signal transduction, apoptosis, oxidative response, processes related with the cytoskeleton and structure, recognition and wound healing were identified as representatives of each haemocyte type. Important signalling pathways in the immune response such as MAPK, Ras and NF-κβ seemed to be more relevant for granulocytes, while the Wnt signalling pathway, particularly relevant for wound healing, more relevant in hyalinocytes. The differences in proteins involved in recognition and in cytoskeleton and structure suggest differential specialisation in processes of phagocytosis and internalisation of pathogens between haemocyte types. Apoptosis seemed more active in granulocytes. The differences in proteins involved in oxidative response also suggest different redox processes in each cell type.
Collapse
Affiliation(s)
- Nuria R de la Ballina
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620, Vilanova de Arousa, Spain
| | - Antonio Villalba
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620, Vilanova de Arousa, Spain; Departamento de Ciencias de la Vida, Universidad de Alcalá, 28871, Alcalá de Henares, Spain; Research Centre for Experimental Marine Biology and Biotechnology (PIE), University of the Basque Country (UPV/EHU), 48620, Plentzia, Spain.
| | - Asunción Cao
- Centro de Investigacións Mariñas (CIMA), Consellería do Mar, Xunta de Galicia, 36620, Vilanova de Arousa, Spain
| |
Collapse
|
26
|
Bettegazzi B, Bellani S, Cattaneo S, Codazzi F, Grohovaz F, Zacchetti D. Gα13 Contributes to LPS-Induced Morphological Alterations and Affects Migration of Microglia. Mol Neurobiol 2021; 58:6397-6414. [PMID: 34529232 DOI: 10.1007/s12035-021-02553-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/26/2021] [Indexed: 11/24/2022]
Abstract
Microglia are the resident immune cells of the CNS that are activated in response to a variety of stimuli. This phenotypical change is aimed to maintain the local homeostasis, also by containing the insults and repair the damages. All these processes are tightly regulated and coordinated and a failure in restoring homeostasis by microglia can result in the development of neuroinflammation that can facilitate the progression of pathological conditions. Indeed, chronic microglia activation is commonly recognized as a hallmark of many neurological disorders, especially at an early stage. Many complex pathways, including cytoskeletal remodeling, are involved in the control of the microglial phenotypical and morphological changes that occur during activation. In this work, we focused on the small GTPase Gα13 and its role at the crossroad between RhoA and Rac1 signaling when microglia is exposed to pro-inflammatory stimulation. We propose the direct involvement of Gα13 in the cytoskeletal rearrangements mediated by FAK, LIMK/cofilin, and Rac1 during microglia activation. In fact, we show that Gα13 knockdown significantly inhibited LPS-induced microglial cell activation, in terms of both changes in morphology and migration, through the modulation of FAK and one of its downstream effectors, Rac1. In conclusion, we propose Gα13 as a critical factor in the regulation of morphological and functional properties of microglia during activation, which might become a target of intervention for the control of microglia inflammation.
Collapse
Affiliation(s)
- Barbara Bettegazzi
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy.
| | - Serena Bellani
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Stefano Cattaneo
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Franca Codazzi
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Fabio Grohovaz
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Daniele Zacchetti
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
27
|
Shen J, Zhao M, Zhang C, Sun X. IL-1β in atherosclerotic vascular calcification: From bench to bedside. Int J Biol Sci 2021; 17:4353-4364. [PMID: 34803503 PMCID: PMC8579452 DOI: 10.7150/ijbs.66537] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/11/2021] [Indexed: 01/19/2023] Open
Abstract
Atherosclerotic vascular calcification contributes to increased risk of death in patients with cardiovascular diseases. Assessing the type and severity of inflammation is crucial in the treatment of numerous cardiovascular conditions. IL-1β, a potent proinflammatory cytokine, plays diverse roles in the pathogenesis of atherosclerotic vascular calcification. Several large-scale, population cohort trials have shown that the incidence of cardiovascular events is clinically reduced by the administration of anti-IL-1β therapy. Anti-IL-1β therapy might reduce the incidence of cardiovascular events by affecting atherosclerotic vascular calcification, but the mechanism underlying this effect remains unclear. In this review, we summarize current knowledge on the role of IL-1β in atherosclerotic vascular calcification, and describe the latest results reported in clinical trials evaluating anti-IL-1β therapies for the treatment of cardiovascular diseases. This review will aid in improving current understanding of the pathophysiological roles of IL-1β and mechanisms underlying its activity.
Collapse
Affiliation(s)
- Jialing Shen
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ming Zhao
- Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chunxiang Zhang
- Laboratory of Nucleic Acids in Medicine for National high-level talents, Southwest Medical University, Luzhou 646000, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Xiaolei Sun
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Laboratory of Nucleic Acids in Medicine for National high-level talents, Southwest Medical University, Luzhou 646000, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King's College London, London SE5 9NU, United Kingdom.,Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou 646000, China
| |
Collapse
|
28
|
Bekere I, Huang J, Schnapp M, Rudolph M, Berneking L, Ruckdeschel K, Grundhoff A, Günther T, Fischer N, Aepfelbacher M. Yersinia remodels epigenetic histone modifications in human macrophages. PLoS Pathog 2021; 17:e1010074. [PMID: 34793580 PMCID: PMC8639070 DOI: 10.1371/journal.ppat.1010074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 10/28/2021] [Indexed: 01/10/2023] Open
Abstract
Various pathogens systematically reprogram gene expression in macrophages, but the underlying mechanisms are largely unknown. We investigated whether the enteropathogen Yersinia enterocolitica alters chromatin states to reprogram gene expression in primary human macrophages. Genome-wide chromatin immunoprecipitation (ChIP) seq analyses showed that pathogen-associated molecular patterns (PAMPs) induced up- or down-regulation of histone modifications (HMod) at approximately 14500 loci in promoters and enhancers. Effectors of Y. enterocolitica reorganized about half of these dynamic HMod, with the effector YopP being responsible for about half of these modulatory activities. The reorganized HMod were associated with genes involved in immune response and metabolism. Remarkably, the altered HMod also associated with 61% of all 534 known Rho GTPase pathway genes, revealing a new level in Rho GTPase regulation and a new aspect of bacterial pathogenicity. Changes in HMod were associated to varying degrees with corresponding gene expression, e. g. depending on chromatin localization and cooperation of the HMod. In summary, infection with Y. enterocolitica remodels HMod in human macrophages to modulate key gene expression programs of the innate immune response. Human pathogenic bacteria can affect epigenetic histone modifications to modulate gene expression in host cells. However, a systems biology analysis of this bacterial virulence mechanism in immune cells has not been performed. Here we analyzed genome-wide epigenetic histone modifications and associated gene expression changes in primary human macrophages infected with enteropathogenic Yersinia enterocolitica. We demonstrate that Yersinia virulence factors extensively modulate histone modifications and associated gene expression triggered by the pathogen-associated molecular patterns (PAMPs) of the bacteria. The epigenetically modulated genes are involved in several key pathways of the macrophage immune response, including the Rho GTPase pathway, revealing a novel level of Rho GTPase regulation by a bacterial pathogen. Overall, our findings provide an in-depth view of epigenetic and gene expression changes during host-pathogen interaction and might have further implications for understanding of the innate immune memory in macrophages.
Collapse
Affiliation(s)
- Indra Bekere
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- * E-mail: (IB); (MA)
| | - Jiabin Huang
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Marie Schnapp
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Maren Rudolph
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Laura Berneking
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Klaus Ruckdeschel
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Adam Grundhoff
- Heinrich-Pette-Institute (HPI), Leibniz Institute for Experimental Virology, Research Group Virus Genomics, Hamburg, Germany
| | - Thomas Günther
- Heinrich-Pette-Institute (HPI), Leibniz Institute for Experimental Virology, Research Group Virus Genomics, Hamburg, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- * E-mail: (IB); (MA)
| |
Collapse
|
29
|
MIDESP: Mutual Information-Based Detection of Epistatic SNP Pairs for Qualitative and Quantitative Phenotypes. BIOLOGY 2021; 10:biology10090921. [PMID: 34571798 PMCID: PMC8469369 DOI: 10.3390/biology10090921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary The interactions between SNPs, which are known as epistasis, can strongly influence the phenotype. Their detection is still a challenge, which is made even more difficult through the existence of background associations that can hide correct epistatic interactions. To address the limitations of existing methods, we present in this study our novel method MIDESP for the detection of epistatic SNP pairs. It is the first mutual information-based method that can be applied to both qualitative and quantitative phenotypes and which explicitly accounts for background associations in the dataset. Abstract The interactions between SNPs result in a complex interplay with the phenotype, known as epistasis. The knowledge of epistasis is a crucial part of understanding genetic causes of complex traits. However, due to the enormous number of SNP pairs and their complex relationship to the phenotype, identification still remains a challenging problem. Many approaches for the detection of epistasis have been developed using mutual information (MI) as an association measure. However, these methods have mainly been restricted to case–control phenotypes and are therefore of limited applicability for quantitative traits. To overcome this limitation of MI-based methods, here, we present an MI-based novel algorithm, MIDESP, to detect epistasis between SNPs for qualitative as well as quantitative phenotypes. Moreover, by incorporating a dataset-dependent correction technique, we deal with the effect of background associations in a genotypic dataset to separate correct epistatic interaction signals from those of false positive interactions resulting from the effect of single SNP×phenotype associations. To demonstrate the effectiveness of MIDESP, we apply it on two real datasets with qualitative and quantitative phenotypes, respectively. Our results suggest that by eliminating the background associations, MIDESP can identify important genes, which play essential roles for bovine tuberculosis or the egg weight of chickens.
Collapse
|
30
|
Witas R, Rasmussen A, Scofield RH, Radfar L, Stone DU, Grundahl K, Lewis D, Sivils KL, Lessard CJ, Farris AD, Nguyen CQ. Defective Efferocytosis in a Murine Model of Sjögren's Syndrome Is Mediated by Dysfunctional Mer Tyrosine Kinase Receptor. Int J Mol Sci 2021; 22:9711. [PMID: 34575873 PMCID: PMC8466327 DOI: 10.3390/ijms22189711] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Sjögren's syndrome (SjS) is a chronic autoimmune disease primarily involving the exocrine glands in which the involvement of the innate immune system is largely uncharacterized. Mer signaling has been found to be protective in several autoimmune diseases but remains unstudied in SjS. Here, we investigated the role of Mer signaling in SjS. Mer knockout (MerKO) mice were examined for SjS disease criteria. SjS-susceptible (SjSS) C57BL/6.NOD-Aec1Aec2 mice were assessed for defective Mer signaling outcomes, soluble Mer (sMer) levels, A disintegrin and metalloprotease 17 (ADAM17) activity, and Rac1 activation. In addition, SjS patient plasma samples were evaluated for sMer levels via ELISA, and sMer levels were correlated to disease manifestations. MerKO mice developed submandibular gland (SMG) lymphocytic infiltrates, SMG apoptotic cells, anti-nuclear autoantibodies (ANA), and reduced saliva flow. Mer signaling outcomes were observed to be diminished in SjSS mice, as evidenced by reduced Rac1 activation in SjSS mice macrophages in response to apoptotic cells and impaired efferocytosis. Increased sMer was also detected in SjSS mouse sera, coinciding with higher ADAM17 activity, the enzyme responsible for cleavage and inactivation of Mer. sMer levels were elevated in patient plasma and positively correlated with focus scores, ocular staining scores, rheumatoid factors, and anti-Ro60 levels. Our data indicate that Mer plays a protective role in SjS, similar to other autoimmune diseases. Furthermore, we suggest a series of events where enhanced ADAM17 activity increases Mer inactivation and depresses Mer signaling, thus removing protection against the loss of self-tolerance and the onset of autoimmune disease in SjSS mice.
Collapse
Affiliation(s)
- Richard Witas
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32608, USA
| | - Astrid Rasmussen
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (A.R.); (K.G.); (C.J.L.)
| | - Robert H. Scofield
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.H.S.); (K.L.S.); (A.D.F.)
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Lida Radfar
- Department of Oral Diagnosis and Radiology, College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Donald U. Stone
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Kiely Grundahl
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (A.R.); (K.G.); (C.J.L.)
| | - David Lewis
- Department of Oral Pathology, College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Kathy L. Sivils
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.H.S.); (K.L.S.); (A.D.F.)
| | - Christopher J. Lessard
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (A.R.); (K.G.); (C.J.L.)
| | - A. Darise Farris
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (R.H.S.); (K.L.S.); (A.D.F.)
| | - Cuong Q. Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32608, USA
- Center of Orphaned Autoimmune Diseases, University of Florida, Gainesville, FL 32611-0880, USA
| |
Collapse
|
31
|
Almutairi O, Almutairi HA, Rushood MA. Protein-Activated Kinase 3 (PAK3)-Related Intellectual Disability Associated with Combined Immunodeficiency: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e930966. [PMID: 34014906 PMCID: PMC8147901 DOI: 10.12659/ajcr.930966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/14/2021] [Accepted: 03/25/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND X-linked intellectual disabilities constitute a group of clinically and genetically heterogeneous disorders that are divided into syndromic and nonsyndromic forms. PAK3 mutations are associated with X-linked nonsyndromic forms of intellectual disability, with the most common clinical features being cognitive deficit, large ears, oral motor hypotonia, and neurobehavioral abnormalities. These mutations have been reported to be associated with either loss of the PAK3 protein or loss of its kinase activity. We report a case with the novel PAK3 variant c.685C>T p.(Pro229Ser), which has not been previously described. CASE REPORT We report the first case of a PAK3 mutation to present with the common clinical features along with immunodeficiency resembling common variable immune deficiency. Our patient was a 10-year-old girl who had experienced septic shock with a rapidly deteriorating course when she was 5-years-old. The initial immune work-up showed lymphopenia affecting all cell lines, but preferentially the B-cell compartment. Further work-up of this patient revealed low levels of immunoglobulin (Ig) G, undetectable IgA, reduced IgG1 and IgG2 subclasses, and poor response to the diphtheria/tetanus vaccine. Lymphocyte function, tested as the response to the mitogen phytohemagglutinin, was low and fluctuated between 9% and 22% compared with control samples. The patient experienced recurrent respiratory tract infections, and she responded well to regular intravenous Ig treatment and antibiotic prophylaxis. CONCLUSIONS The current case might provide a new insight into PAK3 gene function. Although further evidence is needed, it is worth considering that immunological abnormalities may be associated with PAK3 gene mutations.
Collapse
Affiliation(s)
| | | | - Maysoun Al Rushood
- Department of Pediatrics, Faculty of Medicine, Health Sciences Center, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
32
|
Yousefnezhad S, Gharesouran J, Ghafouri-Fard S, Hosseinzadeh H, Ahmadian-Heris J, Jafari-Rouhi AH, Taheri M, Rezazadeh M. DOCK8-related Immunodeficiency Syndrome (DIDS): Report of Novel Mutations in Iranian Patients. J Mol Neurosci 2021; 71:2456-2461. [PMID: 33948880 DOI: 10.1007/s12031-021-01843-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
DOCK8 immunodeficiency syndrome (DIDS) is a rare autosomal recessive (AR) disorder characterized by elevated serum IgE levels, eosinophilia, recurrent cutaneous infections, severe eczema, and sinopulmonary and gastrointestinal infections. This syndrome is a multisystem disease that is associated with both immune deficiency and neurological complications. In this study, we describe the clinical characteristics of two Iranian patients with DOCK8 deficiency and propose possible mechanisms for this condition. By using whole exome sequencing (WES), we identified two novel mutations, namely c.3233_3234del AG (p.Q1078fs) in exon 6 and a large deletion with 94 kb (c.405-3231 deletion, p.K135fs), in these two patients. These variations are confirmed with Sanger sequencing and CGH array. Subsequent co-segregation analysis is performed to identify inheritance patterns. Both patients were homozygote and their parents were heterozygote for the variations. For further investigation, prediction tools were applied to identify the pathogenicity of the variations and also for modeling the truncated proteins. The patients did not show neurological abnormalities associated with a deficiency of the N terminal region of DOCK8. The absence of neurological complications in the first patient is justifiable due to the maintenance of the proline-rich region in DOCK8, but for the second patient with expanded deletion which is almost like null DOCK8 protein, it is not presumable, pointing to the fact that the C terminal region of the protein might have functions in the proliferation and migration neurons in the peripheral nervous system. Alternatively, it is possible that neurological abnormalities follow an age-dependent pattern, leading to the appearance of related symptoms later in life. Further multiple functional studies are needed to model different identified variants in animal models to confirm our results and suggest possible mechanisms associated with DOCK8 deficiency in this study.
Collapse
Affiliation(s)
| | - Jalal Gharesouran
- Molecular Genetics Division, GMG center, Tabriz, Iran.,Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Hosseinzadeh
- Molecular Genetics Division, GMG center, Tabriz, Iran.,Department of Allergy and Clinical Immunology, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian-Heris
- Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Rezazadeh
- Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Abstract
Pathogens have evolved smart strategies to invade hosts and hijack their immune responses. One such strategy is the targeting of the host RhoGTPases by toxins or virulence factors to hijack the cytoskeleton dynamic and immune processes. In response to this microbial attack, the host has evolved an elegant strategy to monitor the function of virulence factors and toxins by sensing the abnormal activity of RhoGTPases. This innate immune strategy of sensing bacterial effector targeting RhoGTPase appears to be a bona fide example of effector-triggered immunity (ETI). Here, we review recently discovered mechanisms by which the host can sense the activity of these toxins through NOD and NOD-like receptors (NLRs).
Collapse
Affiliation(s)
| | - Laurent Boyer
- Université Côte d’Azur, Inserm, C3M, Nice, France
- * E-mail:
| |
Collapse
|
34
|
Dong Q, Lunney JK, Lim KS, Nguyen Y, Hess AS, Beiki H, Rowland RRR, Walker K, Reecy JM, Tuggle CK, Dekkers JCM. Gene expression in tonsils in swine following infection with porcine reproductive and respiratory syndrome virus. BMC Vet Res 2021; 17:88. [PMID: 33618723 PMCID: PMC7901068 DOI: 10.1186/s12917-021-02785-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/02/2021] [Indexed: 12/18/2022] Open
Abstract
Background Porcine reproductive and respiratory syndrome (PRRS) is a threat to pig production worldwide. Our objective was to understand mechanisms of persistence of PRRS virus (PRRSV) in tonsil. Transcriptome data from tonsil samples collected at 42 days post infection (dpi) were generated by RNA-seq and NanoString on 51 pigs that were selected to contrast the two PRRSV isolates used, NVSL and KS06, high and low tonsil viral level at 42 dpi, and the favorable and unfavorable genotypes at a genetic marker (WUR) for the putative PRRSV resistance gene GBP5. Results The number of differentially expressed genes (DEGs) differed markedly between models with and without accounting for cell-type enrichments (CE) in the samples that were predicted from the RNA-seq data. This indicates that differences in cell composition in tissues that consist of multiple cell types, such as tonsil, can have a large impact on observed differences in gene expression. Based on both the NanoString and the RNA-seq data, KS06-infected pigs showed greater activation, or less inhibition, of immune response in tonsils at 42 dpi than NVSL-infected pigs, with and without accounting for CE. This suggests that the NVSL virus may be better than the KS06 virus at evading host immune response and persists in tonsils by weakening, or preventing, host immune responses. Pigs with high viral levels showed larger CE of immune cells than low viral level pigs, potentially to trigger stronger immune responses. Presence of high tonsil virus was associated with a stronger immune response, especially innate immune response through interferon signaling, but these differences were not significant when accounting for CE. Genotype at WUR was associated with different effects on immune response in tonsils of pigs during the persistence stage, depending on viral isolate and tonsil viral level. Conclusions Results of this study provide insights into the effects of PRRSV isolate, tonsil viral level, and WUR genotype on host immune response and into potential mechanisms of PRRSV persistence in tonsils that could be targeted to improve strategies to reduce viral rebreaks. Finally, to understand transcriptome responses in tissues that consist of multiple cell types, it is important to consider differences in cell composition. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-021-02785-1.
Collapse
Affiliation(s)
- Qian Dong
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | | | - Kyu-Sang Lim
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | - Yet Nguyen
- Department of Statistics, Iowa State University, Ames, Iowa, 50011, USA
| | - Andrew S Hess
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | - Hamid Beiki
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | - Raymond R R Rowland
- College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | | | - James M Reecy
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | | | - Jack C M Dekkers
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
35
|
Functional and Therapeutic Relevance of Rho GTPases in Innate Immune Cell Migration and Function during Inflammation: An In Silico Perspective. Mediators Inflamm 2021; 2021:6655412. [PMID: 33628114 PMCID: PMC7896857 DOI: 10.1155/2021/6655412] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Systematic regulation of leukocyte migration to the site of infection is a vital step during immunological responses. Improper migration and localization of immune cells could be associated with disease pathology as seen in systemic inflammation. Rho GTPases act as molecular switches during inflammatory cell migration by cycling between Rho-GDP (inactive) to Rho-GTP (active) forms and play an essential role in the precise regulation of actin cytoskeletal dynamics as well as other immunological functions of leukocytes. Available reports suggest that the dysregulation of Rho GTPase signaling is associated with various inflammatory diseases ranging from mild to life-threatening conditions. Therefore, it is crucial to understand the step-by-step activation and inactivation of GTPases and the functioning of different Guanine Nucleotide Exchange Factors (GEFs) and GTPase-Activating Proteins (GAPs) that regulate the conversion of GDP to GTP and GTP to GDP exchange reactions, respectively. Here, we describe the molecular organization and activation of various domains of crucial elements associated with the activation of Rho GTPases using solved PDB structures. We will also present the latest evidence available on the relevance of Rho GTPases in the migration and function of innate immune cells during inflammation. This knowledge will help scientists design promising drug candidates against the Rho-GTPase-centric regulatory molecules regulating inflammatory cell migration.
Collapse
|
36
|
Gao L, Kong L, Zhao Y. The Regulatory Role of Rho GTPases and their Substrates in Osteoclastogenesis. Curr Drug Targets 2021; 22:1064-1070. [PMID: 32981499 DOI: 10.2174/1389450121666200925150446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/15/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
Abstract
Pathological bone loss diseases (osteolysis, Paget's diseases) are commonly caused by the excessive differentiation and activity of osteoclasts. The Rho GTPases family members Rac1/2 (Rac1 and Rac2) have been reported for their special role in exerting multiple cellular functions during osteoclastic differentiation, which includes the most prominent function on dynamic actin cytoskeleton rearranging. Besides that, the increasing studies demonstrated that the regulating effects of Rac1/2 on the osteoclastic cytoskeletal organization are through the GEFs member Dock5. Although the amount of relevant studies on this topic is still limited, several excellent studies have been reported that extensively explored the molecular mechanisms involved in Rac1/2 and Dock5 during the osteoclastogenesis regulation, as well as their role as the therapeutic target in bone loss diseases. Herein, in this review, we aim to focus on recent advances studies for extensively understanding the role of Rho GTPases Rac1/2 and Dock5 in osteoclastogenesis, as well as their role as a potential therapeutic target in regulating osteoclastogenesis.
Collapse
Affiliation(s)
- Lin Gao
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi'an Jiaotong University, China
| | - Lingbo Kong
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi'an Jiaotong University, China
| | - Yuanting Zhao
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi'an Jiaotong University, China
| |
Collapse
|
37
|
Ungefroren H, Wellner UF, Keck T, Lehnert H, Marquardt JU. The Small GTPase RAC1B: A Potent Negative Regulator of-and Useful Tool to Study-TGFβ Signaling. Cancers (Basel) 2020; 12:E3475. [PMID: 33266416 PMCID: PMC7700615 DOI: 10.3390/cancers12113475] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
RAC1 and its alternatively spliced isoform, RAC1B, are members of the Rho family of GTPases. Both isoforms are involved in the regulation of actin cytoskeleton remodeling, cell motility, cell proliferation, and epithelial-mesenchymal transition (EMT). Compared to RAC1, RAC1B exhibits a number of distinctive features with respect to tissue distribution, downstream signaling and a role in disease conditions like inflammation and cancer. The subcellular locations and interaction partners of RAC1 and RAC1B vary depending on their activation state, which makes RAC1 and RAC1B ideal candidates to establish cross-talk with cancer-associated signaling pathways-for instance, interactions with signaling by transforming growth factor β (TGFβ), a known tumor promoter. Although RAC1 has been found to promote TGFβ-driven tumor progression, recent observations in pancreatic carcinoma cells surprisingly revealed that RAC1B confers anti-oncogenic properties, i.e., through inhibiting TGFβ-induced EMT. Since then, an unexpected array of mechanisms through which RAC1B cross-talks with TGFβ signaling has been demonstrated. However, rather than being uniformly inhibitory, RAC1B interacts with TGFβ signaling in a way that results in the selective blockade of tumor-promoting pathways, while concomitantly allowing tumor-suppressive pathways to proceed. In this review article, we are going to discuss the specific interactions between RAC1B and TGFβ signaling, which occur at multiple levels and include various components such as ligands, receptors, cytosolic mediators, transcription factors, and extracellular inhibitors of TGFβ ligands.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany;
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, Campus Kiel, University Hospital Schleswig-Holstein, D-24105 Kiel, Germany
| | - Ulrich F. Wellner
- Clinic for Surgery, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany; (U.F.W.); (T.K.)
| | - Tobias Keck
- Clinic for Surgery, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany; (U.F.W.); (T.K.)
| | | | - Jens-Uwe Marquardt
- First Department of Medicine, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany;
| |
Collapse
|
38
|
Regulation of Metabolic Processes by Hydrogen Peroxide Generated by NADPH Oxidases. Processes (Basel) 2020. [DOI: 10.3390/pr8111424] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hydrogen peroxide (H2O2) is an important oxidizing molecule that regulates the metabolisms of aerobic organisms. Redox signaling comprises physiological oxidative stress (eustress), while excessive oxidative stress causes damage to molecules. The main enzymatic generators of H2O2 are nicotinamide adenine dinucleotide phosphate oxidases or NADPH oxidases (NOXs) and mitochondrial respiratory chains, as well as various oxidases. The NOX family is constituted of seven enzyme isoforms that produce a superoxide anion (O2−), which can be converted to H2O2 by superoxide dismutase or spontaneously. H2O2 passes through the membranes by some aquaporins (AQPs), known as peroxyporins. It diffuses through cells and tissues to initiate cellular effects, such as proliferation, the recruitment of immune cells, and cell shape changes. Therefore, it has been proposed that H2O2 has the same importance as Ca2+ or adenosine triphosphate (ATP) to act as modulators in signaling and the metabolism. The present overview focuses on the metabolic processes of liver and adipose tissue, regulated by the H2O2 generated by NOXs.
Collapse
|
39
|
Sadri S, Tomar N, Yang C, Audi SH, Cowley AW, Dash RK. Mechanistic computational modeling of the kinetics and regulation of NADPH oxidase 2 assembly and activation facilitating superoxide production. Free Radic Res 2020; 54:695-721. [PMID: 33059489 DOI: 10.1080/10715762.2020.1836368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species (ROS) play a crucial role in many physiological processes. However, ROS overproduction leads to oxidative stress, which plays a critical role in cell injury/death and the pathogenesis of many diseases. Members of NADPH oxidase (NOX) family, most of which are comprised of membrane and cytosolic components, are known to be the major nonmitochondrial sources of ROS in many cells. NOX2 is a widely-expressed and well-studied NOX family member, which is activated upon assembly of its membrane subunits gp91 phox and p22 phox with its cytosolic subunits p40 phox , p47 phox , p67 phox , and Rac, facilitating ROS production. NOX2 activation is also enhanced by GTP and inhibited by GDP. However, there remains a lack of a mechanistic, quantitative, and integrated understanding of the kinetics and regulation of the assembly of these subunits and their relative contributions toward NOX2 activation and ROS production. Toward this end, we have developed a mechanistic computational model, which incorporates a generalized random rapid equilibrium binding mechanism for NOX2 assembly and activation as well as regulations by GTP (activation), GDP (inhibition), and individual subunits enhancing the binding of other subunits (mutual binding enhancement). The resulting model replicates diverse published kinetic data, including subunit concentration-dependent NOX2 activation and ROS production, under different assay conditions, with appropriate estimates of the unknown model parameters. The model provides a mechanistic, quantitative, and integrated framework for investigating the critical roles of NOX2 subunits in NOX2 assembly and activation facilitating ROS production in a variety of physiological and pathophysiological conditions. However, there is also a need for better quantitative kinetic data based on current understanding of NOX2 assembly and activation in order to test and further develop this model.
Collapse
Affiliation(s)
- Shima Sadri
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WIS, USA
| | - Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WIS, USA
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WIS, USA
| | - Said H Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, WIS, USA
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WIS, USA
| | - Ranjan K Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WIS, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WIS, USA
| |
Collapse
|
40
|
Liu Y, Dou Y, Yan L, Yang X, He B, Kong L, Smith W. The role of Rho GTPases' substrates Rac and Cdc42 in osteoclastogenesis and relevant natural medicinal products study. Biosci Rep 2020; 40:BSR20200407. [PMID: 32578854 PMCID: PMC7364480 DOI: 10.1042/bsr20200407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/07/2020] [Accepted: 06/23/2020] [Indexed: 12/30/2022] Open
Abstract
Recently, Rho GTPases substrates include Rac (Rac1 and Rac2) and Cdc42 that have been reported to exert multiple cellular functions in osteoclasts, the most prominent of which includes regulating the dynamic actin cytoskeleton rearrangements. In addition, natural products and their molecular frameworks have a long tradition as valuable starting points for medicinal chemistry and drug discovery. Although currently, there are reports about the natural product, which could play a therapeutic role in bone loss diseases (osteoporosis and osteolysis) through the regulation of Rac1/2 and Cdc42 during osteoclasts cytoskeletal structuring. There have been several excellent studies for exploring the therapeutic potentials of various natural products for their role in inhibiting cancer cells migration and function via regulating the Rac1/2 and Cdc42. Herein in this review, we try to focus on recent advancement studies for extensively understanding the role of Rho GTPases substrates Rac1, Rac2 and Cdc42 in osteoclastogenesis, as well as therapeutic potentials of natural medicinal products for their properties on the regulation of Rac1, and/or Rac2 and Cdc42, which is in order to inspire drug discovery in regulating osteoclastogenesis.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi’an Jiaotong University, Xi'an, China
- Department of Orthopedics, Yan’an University Medical School, Yan’an, China
| | - Yusheng Dou
- Department of Shoulder and Elbow Joint, Honghui Hospital, School of Medicine, Xi’an Jiaotong University, China
| | - Liang Yan
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi’an Jiaotong University, Xi'an, China
| | - Xiaobin Yang
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi’an Jiaotong University, Xi'an, China
| | - Baorong He
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi’an Jiaotong University, Xi'an, China
| | - Lingbo Kong
- Department of Spine Surgery, Honghui Hospital, School of Medicine, Xi’an Jiaotong University, Xi'an, China
| | - Wanli Smith
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, U.S.A
| |
Collapse
|
41
|
Schwensow N, Pederson S, Peacock D, Cooke B, Cassey P. Adaptive changes in the genomes of wild rabbits after 16 years of viral epidemics. Mol Ecol 2020; 29:3777-3794. [PMID: 32506669 DOI: 10.1111/mec.15498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 05/20/2020] [Accepted: 05/29/2020] [Indexed: 01/01/2023]
Abstract
Since its introduction to control overabundant invasive European rabbits (Oryctolagus cuniculus), the highly virulent rabbit haemorrhagic disease virus (RHDV) has caused regular annual disease outbreaks in Australian rabbit populations. Although initially reducing rabbit abundance by 60%, continent-wide, experimental evidence has since indicated increased genetic resistance in wild rabbits that have experienced RHDV-driven selection. To identify genetic adaptations, which explain the increased resistance to this biocontrol virus, we investigated genome-wide SNP (single nucleotide polymorphism) allele frequency changes in a South Australian rabbit population that was sampled in 1996 (pre-RHD genomes) and after 16 years of RHDV outbreaks. We identified several SNPs with changed allele frequencies within or close to genes potentially important for increased RHD resistance. The identified genes are known to be involved in virus infections and immune reactions or had previously been identified as being differentially expressed in healthy versus acutely RHDV-infected rabbits. Furthermore, we show in a simulation study that the allele/genotype frequency changes cannot be explained by drift alone and that several candidate genes had also been identified as being associated with surviving RHD in a different Australian rabbit population. Our unique data set allowed us to identify candidate genes for RHDV resistance that have evolved under natural conditions, and over a time span that would not have been feasible in an experimental setting. Moreover, it provides a rare example of host genetic adaptations to virus-driven selection in response to a suddenly emerging infectious disease.
Collapse
Affiliation(s)
- Nina Schwensow
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Ulm, Germany.,Centre for Applied Conservation Science, and School of Biological Sciences, University of Adelaide, SA, Australia
| | - Stephen Pederson
- Bioinformatics Hub, School of Biological Sciences, University of Adelaide, SA, Australia
| | - David Peacock
- Biosecurity SA, Adelaide, SA, Australia.,School of Animal and Veterinary Science, University of Adelaide, Roseworthy, SA, Australia
| | - Brian Cooke
- Institute for Applied Ecology, University of Canberra, Canberra, ACT, Australia
| | - Phillip Cassey
- Centre for Applied Conservation Science, and School of Biological Sciences, University of Adelaide, SA, Australia
| |
Collapse
|
42
|
Chen Y, Chen Y, Yin W, Han H, Miller H, Li J, Herrada AA, Kubo M, Sui Z, Gong Q, Liu C. The regulation of DOCK family proteins on T and B cells. J Leukoc Biol 2020; 109:383-394. [PMID: 32542827 DOI: 10.1002/jlb.1mr0520-221rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 01/01/2023] Open
Abstract
The dedicator of cytokinesis (DOCK) family proteins consist of 11 members, each of which contains 2 domains, DOCK homology region (DHR)-1 and DHR-2, and as guanine nucleotide exchange factors, they mediate activation of small GTPases. Both DOCK2 and DOCK8 deficiencies in humans can cause severe combined immunodeficiency, but they have different characteristics. DOCK8 defect mainly causes high IgE, allergic disease, refractory skin virus infection, and increased incidence of malignant tumor, whereas DOCK2 defect mainly causes early-onset, invasive infection with less atopy and increased IgE. However, the underlying molecular mechanisms causing the disease remain unclear. This paper discusses the role of DOCK family proteins in regulating B and T cells, including development, survival, migration, activation, immune tolerance, and immune functions. Moreover, related signal pathways or molecule mechanisms are also described in this review. A greater understanding of DOCK family proteins and their regulation of lymphocyte functions may facilitate the development of new therapeutics for immunodeficient patients and improve their prognosis.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi, Zunyi, Guizhou, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Han
- Department of Hematology of Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- The Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jianrong Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andres A Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomedicas, Universidad Autonoma de Chile, Talca, Chile
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa, Japan
| | - Zhiwei Sui
- Division of Medical and Biological Measurement, National Institute of Metrology, Beijing, China
| | - Quan Gong
- Department of immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Kunimura K, Uruno T, Fukui Y. DOCK family proteins: key players in immune surveillance mechanisms. Int Immunol 2020; 32:5-15. [PMID: 31630188 PMCID: PMC6949370 DOI: 10.1093/intimm/dxz067] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022] Open
Abstract
Dedicator of cytokinesis (DOCK) proteins constitute a family of evolutionarily conserved guanine nucleotide exchange factors (GEFs) for the Rho family of GTPases. Although DOCK family proteins do not contain the Dbl homology domain typically found in other GEFs, they mediate the GTP–GDP exchange reaction through the DOCK homology region-2 (DHR-2) domain. In mammals, this family consists of 11 members, each of which has unique functions depending on the expression pattern and the substrate specificity. For example, DOCK2 is a Rac activator critical for migration and activation of leukocytes, whereas DOCK8 is a Cdc42-specific GEF that regulates interstitial migration of dendritic cells. Identification of DOCK2 and DOCK8 as causative genes for severe combined immunodeficiency syndromes in humans has highlighted their roles in immune surveillance. In addition, the recent discovery of a naturally occurring DOCK2-inhibitory metabolite has uncovered an unexpected mechanism of tissue-specific immune evasion. On the other hand, GEF-independent functions have been shown for DOCK8 in antigen-induced IL-31 production in helper T cells. This review summarizes multifaced functions of DOCK family proteins in the immune system.
Collapse
Affiliation(s)
- Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan.,Research Center for Advanced Immunology, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan.,Research Center for Advanced Immunology, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
44
|
Annunziata MC, Parisi M, Esposito G, Fabbrocini G, Ammendola R, Cattaneo F. Phosphorylation Sites in Protein Kinases and Phosphatases Regulated by Formyl Peptide Receptor 2 Signaling. Int J Mol Sci 2020; 21:ijms21113818. [PMID: 32471307 PMCID: PMC7312799 DOI: 10.3390/ijms21113818] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
FPR1, FPR2, and FPR3 are members of Formyl Peptides Receptors (FPRs) family belonging to the GPCR superfamily. FPR2 is a low affinity receptor for formyl peptides and it is considered the most promiscuous member of this family. Intracellular signaling cascades triggered by FPRs include the activation of different protein kinases and phosphatase, as well as tyrosine kinase receptors transactivation. Protein kinases and phosphatases act coordinately and any impairment of their activation or regulation represents one of the most common causes of several human diseases. Several phospho-sites has been identified in protein kinases and phosphatases, whose role may be to expand the repertoire of molecular mechanisms of regulation or may be necessary for fine-tuning of switch properties. We previously performed a phospho-proteomic analysis in FPR2-stimulated cells that revealed, among other things, not yet identified phospho-sites on six protein kinases and one protein phosphatase. Herein, we discuss on the selective phosphorylation of Serine/Threonine-protein kinase N2, Serine/Threonine-protein kinase PRP4 homolog, Serine/Threonine-protein kinase MARK2, Serine/Threonine-protein kinase PAK4, Serine/Threonine-protein kinase 10, Dual specificity mitogen-activated protein kinase kinase 2, and Protein phosphatase 1 regulatory subunit 14A, triggered by FPR2 stimulation. We also describe the putative FPR2-dependent signaling cascades upstream to these specific phospho-sites.
Collapse
Affiliation(s)
- Maria Carmela Annunziata
- Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.A.); (M.P.); (G.F.)
| | - Melania Parisi
- Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.A.); (M.P.); (G.F.)
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (G.E.); (R.A.)
| | - Gabriella Fabbrocini
- Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.A.); (M.P.); (G.F.)
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (G.E.); (R.A.)
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (G.E.); (R.A.)
- Correspondence: ; Fax: +39-081-7464-359
| |
Collapse
|
45
|
Bankers L, Miller C, Liu G, Thongkittidilok C, Morrison J, Poeschla EM. Development of IFN-Stimulated Gene Expression from Embryogenesis through Adulthood, with and without Constitutive MDA5 Pathway Activation. THE JOURNAL OF IMMUNOLOGY 2020; 204:2791-2807. [PMID: 32277054 DOI: 10.4049/jimmunol.1901421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/26/2020] [Indexed: 11/19/2022]
Abstract
Pathogen-associated molecular patterns (e.g., dsRNA) activate expression of IFN-stimulated genes (ISGs), which protect hosts from infection. Although transient ISG upregulation is essential for effective innate immunity, constitutive activation typically causes harmful autoimmunity in mice and humans, often including severe developmental abnormalities. We have shown that transgenic mice expressing a picornavirus RNA-dependent RNA polymerase (RdRP) outside the viral context (RdRP mice) exhibit constitutive, MDA5-dependent, and quantitatively dramatic upregulation of many ISGs, which confers broad viral infection resistance. Remarkably, RdRP mice never develop autoinflammation, interferonopathy, or other discernible abnormalities. In this study, we used RNA sequencing and other methods to analyze ISG expression across five time points from fetal development to adulthood in wild-type and RdRP mice. In RdRP mice, the proportion of upregulated ISGs increased during development, with the most dramatic induction occurring 2 wk postnatally. The amplified ISG profile is then maintained lifelong. Molecular pathways and biological functions associated with innate immune and IFN signaling are only activated postnatally, suggesting constrained fetal responsiveness to innate immune stimuli. Biological functions supporting replication of viruses are only inhibited postnatally. We further determined that the RdRP is expressed at low levels and that blocking Ifnar1 reverses the amplified ISG transcriptome in adults. In conclusion, the upregulated ISG profile of RdRP mice is mostly triggered early postnatally, is maintained through adulthood, and requires ongoing type I IFN signaling to maintain it. The model provides opportunities to study the systems biology of innate immunity and to determine how sustained ISG upregulation can be compatible with robust health.
Collapse
Affiliation(s)
- Laura Bankers
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Caitlin Miller
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Guoqi Liu
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Chommanart Thongkittidilok
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - James Morrison
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Eric M Poeschla
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| |
Collapse
|
46
|
Namekata K, Guo X, Kimura A, Azuchi Y, Kitamura Y, Harada C, Harada T. Roles of the DOCK-D family proteins in a mouse model of neuroinflammation. J Biol Chem 2020; 295:6710-6720. [PMID: 32241915 DOI: 10.1074/jbc.ra119.010438] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/16/2020] [Indexed: 02/01/2023] Open
Abstract
The DOCK-D (dedicator of cytokinesis D) family proteins are atypical guanine nucleotide exchange factors that regulate Rho GTPase activity. The family consists of Zizimin1 (DOCK9), Zizimin2 (DOCK11), and Zizimin3 (DOCK10). Functions of the DOCK-D family proteins are presently not well-explored, and the role of the DOCK-D family in neuroinflammation is unknown. In this study, we generated three mouse lines in which DOCK9 (DOCK9 -/-), DOCK10 (DOCK10 -/-), or DOCK11 (DOCK11 -/-) had been deleted and examined the phenotypic effects of these gene deletions in MOG35-55 peptide-induced experimental autoimmune encephalomyelitis, an animal model of the neuroinflammatory disorder multiple sclerosis. We found that all the gene knockout lines were healthy and viable. The only phenotype observed under normal conditions was a slightly smaller proportion of B cells in splenocytes in DOCK10 -/- mice than in the other mouse lines. We also found that the migration ability of macrophages is impaired in DOCK10 -/- and DOCK11 -/- mice and that the severity of experimental autoimmune encephalomyelitis was ameliorated only in DOCK10 -/- mice. No apparent phenotype was observed for DOCK9 -/- mice. Further investigations indicated that lipopolysaccharide stimulation up-regulates DOCK10 expression in microglia and that microglial migration is decreased in DOCK10 -/- mice. Up-regulation of C-C motif chemokine ligand 2 (CCL2) expression induced by activation of Toll-like receptor 4 or 9 signaling was reduced in DOCK10 -/- astrocytes compared with WT astrocytes. Taken together, our findings suggest that DOCK10 plays a role in innate immunity and neuroinflammation and might represent a potential therapeutic target for managing multiple sclerosis.
Collapse
Affiliation(s)
- Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuriko Azuchi
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuta Kitamura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
47
|
Chen S, Gong Z, Letcher RJ, Liu C. Promotion effect of liver tumor progression in male kras transgenic zebrafish induced by tris (1, 3-dichloro-2-propyl) phosphate. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 191:110220. [PMID: 31991394 DOI: 10.1016/j.ecoenv.2020.110220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 06/10/2023]
Abstract
A previous study reported that exposure to tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) could promote the progression of hepatocellular carcinoma (HCC) in female HCC model zebrafish. Due to the existence of gender disparity in the development of HCC between females and males, whether the promotion effect of TDCIPP still exists in male HCC model zebrafish remains unclear. In this study, Tg(fabp10:rtTA2s-M2; TRE2:EGFP-krasG12V), referred as kras transgenic zebrafish which was shown to be an inducible liver tumor model, was applied as experimental model to assess the promotion potential of TDCIPP for HCC in males. In brief, kras males were exposed to 20 mg/L doxycycline (DOX), 0.3 mg/L TDCIPP and a binary mixture of 20 mg/L DOX with 0.3 mg/L TDCIPP, and after exposure liver size, histopathology and transcriptional profiles of liver from these treatments were examined. With the involvement of TDCIPP, the liver size was significantly increased and the lesion of hepatocyte became more aggressive. Furthermore, expressions of genes involved in DNA replication and inflammatory response were simultaneously up-regulated in the treatment of TDCIPP compared with the solvent control and in the treatment of the binary mixture of the two chemicals compared to the single DOX treatment. Overall, our results suggested that TDCIPP had promotion effect on the progression of liver tumor in kras males.
Collapse
Affiliation(s)
- Sheng Chen
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore
| | - Robert J Letcher
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University, Ottawa, Ontario, K1A 0H3, Canada
| | - Chunsheng Liu
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
48
|
The dual role of curcumin and ferulic acid in counteracting chemoresistance and cisplatin-induced ototoxicity. Sci Rep 2020; 10:1063. [PMID: 31974389 PMCID: PMC6978317 DOI: 10.1038/s41598-020-57965-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Platinum-based agents, such as cisplatin, form the mainstay of currently used chemotherapeutic regimens for several malignancies; however, the main limitations are chemoresistance and ototoxic side effects. In this study we used two different polyphenols, curcumin and ferulic acid as adjuvant chemotherapeutics evaluating (1) in vivo their antioxidant effects in protecting against cisplatin ototoxicity and (2) in vitro the transcription factors involved in tumor progression and cisplatin resistance. We reported that both polyphenols show antioxidant and oto-protective activity in the cochlea by up-regulating Nrf-2/HO-1 pathway and downregulating p53 phosphorylation. However, only curcumin is able to influence inflammatory pathways counteracting NF-κB activation. In human cancer cells, curcumin converts the anti-oxidant effect into a pro-oxidant and anti-inflammatory one. Curcumin exerts permissive and chemosensitive properties by targeting the cisplatin chemoresistant factors Nrf-2, NF-κB and STAT-3 phosphorylation. Ferulic acid shows a biphasic response: it is pro-oxidant at lower concentrations and anti-oxidant at higher concentrations promoting chemoresistance. Thus, polyphenols, mainly curcumin, targeting ROS-modulated pathways may be a promising tool for cancer therapy. Thanks to their biphasic activity of antioxidant in normal cells undergoing stressful conditions and pro-oxidant in cancer cells, these polyphenols probably engage an interplay among the key factors Nrf-2, NF-κB, STAT-3 and p53.
Collapse
|
49
|
Caffo L, Sneed BL, Burcham C, Reed K, Hahn NC, Bell S, Downham O, Evans MD, Fullenkamp CR, Drinnon TK, Bishop D, Bruns HA, McKillip JL, Sammelson RE, McDowell SA. Simvastatin and ML141 Decrease Intracellular Streptococcus pyogenes Infection. Curr Pharm Biotechnol 2019; 20:733-744. [PMID: 31258074 DOI: 10.2174/1389201020666190618115154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/20/2019] [Accepted: 05/14/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Recurrent pharyngotonsillitis due to Streptococcus pyogenes develops regardless of whether infecting strains are resistant or susceptible to first-line antimicrobials. Causation for recurrent infection is associated with the use of first-line antimicrobials that fail to penetrate deep tissue and host cell membranes, enabling intracellular S. pyogenes to survive throughout repeated rounds of antimicrobial therapy. OBJECTIVE To determine whether simvastatin, a therapeutic approved for use in the treatment of hypercholesterolemia, and ML141, a first-in-class small molecule inhibitor with specificity for human CDC42, limit host cell invasion by S. pyogenes. METHODS Assays to assess host cell invasion, bactericidal activity, host cell viability, actin depolymerization, and fibronectin binding were performed using the RAW 267.4 macrophage cell line and Human Umbilical Vein Endothelial Cells (HUVEC) infected with S. pyogenes (90-226) and treated with simvastatin, ML141, structural analogs of ML141, or vehicle control. RESULTS Simvastatin and ML141 decreased intracellular infection by S. pyogenes in a dose-dependent manner. Inhibition by simvastatin persisted following 1 h washout whereas inhibition by ML141 was reversed. During S. pyogenes infection, actin stress fibers depolymerized in vehicle control treated cells, yet remained intact in simvastatin and in ML141 treated cells. Consistent with the previous characterization of ML141, simvastatin decreased host cell binding to fibronectin. Structural analogs of ML141, designated as the RSM series, decreased intracellular infection through non-cytotoxic, nonbactericidal mechanisms. CONCLUSION Our findings demonstrate the potential of repurposing simvastatin and of developing CDC42-targeted therapeutics for eradicating intracellular S. pyogenes infection to break the cycle of recurrent infection through a host-directed approach.
Collapse
Affiliation(s)
- Lindy Caffo
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Bria L Sneed
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Caroline Burcham
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Katie Reed
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Nathan C Hahn
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Samantha Bell
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Olivia Downham
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Melissa D Evans
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | | | - Teague K Drinnon
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Derron Bishop
- Indiana University School of Medicine - Muncie Campus, Muncie, IN, 47306, United States
| | - Heather A Bruns
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - John L McKillip
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Robert E Sammelson
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| | - Susan A McDowell
- Ball State University, RH 105, 2100 West Riverside Avenue, Muncie, Indiana 47306, United States
| |
Collapse
|
50
|
Reactive Oxygen Species Are Key Mediators of Demyelination in Canine Distemper Leukoencephalitis but not in Theiler's Murine Encephalomyelitis. Int J Mol Sci 2019; 20:ijms20133217. [PMID: 31262031 PMCID: PMC6651464 DOI: 10.3390/ijms20133217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 01/04/2023] Open
Abstract
(1) Background: Canine distemper virus (CDV)-induced demyelinating leukoencephalitis (CDV-DL) in dogs and Theiler’s murine encephalomyelitis (TME) virus (TMEV)-induced demyelinating leukomyelitis (TMEV-DL) are virus-induced demyelinating conditions mimicking Multiple Sclerosis (MS). Reactive oxygen species (ROS) can induce the degradation of lipids and nucleic acids to characteristic metabolites such as oxidized lipids, malondialdehyde, and 8-hydroxyguanosine. The hypothesis of this study is that ROS are key effector molecules in the pathogenesis of myelin membrane breakdown in CDV-DL and TMEV-DL. (2) Methods: ROS metabolites and antioxidative enzymes were assessed using immunofluorescence in cerebellar lesions of naturally CDV-infected dogs and spinal cord tissue of TMEV-infected mice. The transcription of selected genes involved in ROS generation and detoxification was analyzed using gene-expression microarrays in CDV-DL and TMEV-DL. (3) Results: Immunofluorescence revealed increased amounts of oxidized lipids, malondialdehyde, and 8-hydroxyguanosine in CDV-DL while TMEV-infected mice did not reveal marked changes. In contrast, microarray-analysis showed an upregulated gene expression associated with ROS generation in both diseases. (4) Conclusion: In summary, the present study demonstrates a similar upregulation of gene-expression of ROS generation in CDV-DL and TMEV-DL. However, immunofluorescence revealed increased accumulation of ROS metabolites exclusively in CDV-DL. These results suggest differences in the pathogenesis of demyelination in these two animal models.
Collapse
|